1
|
Troop LD, Puetzer JL. Intermittent cyclic stretch of engineered ligaments drives hierarchical collagen fiber maturation in a dose- and organizational-dependent manner. Acta Biomater 2024; 185:296-311. [PMID: 39025395 PMCID: PMC11381169 DOI: 10.1016/j.actbio.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Hierarchical collagen fibers are the primary source of strength in tendons and ligaments; however, these fibers largely do not regenerate after injury or with repair, resulting in limited treatment options. We previously developed a static culture system that guides ACL fibroblasts to produce native-sized fibers and early fascicles by 6 weeks. These constructs are promising ligament replacements, but further maturation is needed. Mechanical cues are critical for development in vivo and in engineered tissues; however, the effect on larger fiber and fascicle formation is largely unknown. Our objective was to investigate whether intermittent cyclic stretch, mimicking rapid muscle activity, drives further maturation in our system to create stronger engineered replacements and to explore whether cyclic loading has differential effects on cells at different degrees of collagen organization to better inform engineered tissue maturation protocols. Constructs were loaded with an established intermittent cyclic loading regime at 5 or 10 % strain for up to 6 weeks and compared to static controls. Cyclic loading drove cells to increase hierarchical collagen organization, collagen crimp, and tissue tensile properties, ultimately producing constructs that matched or exceeded immature ACL properties. Further, the effect of loading on cells varied depending on degree of organization. Specifically, 10 % load drove early improvements in tensile properties and composition, while 5 % load was more beneficial later in culture, suggesting a shift in mechanotransduction. This study provides new insight into how cyclic loading affects cell-driven hierarchical fiber formation and maturation, which will help to develop better rehabilitation protocols and engineer stronger replacements. STATEMENT OF SIGNIFICANCE: Collagen fibers are the primary source of strength and function in tendons and ligaments throughout the body. These fibers have limited regenerate after injury, with repair, and in engineered replacements, reducing treatment options. Cyclic load has been shown to improve fibril level alignment, but its effect at the larger fiber and fascicle length-scale is largely unknown. Here, we demonstrate intermittent cyclic loading increases cell-driven hierarchical fiber formation and tissue mechanics, producing engineered replacements with similar organization and mechanics as immature ACLs. This study provides new insight into how cyclic loading affects cell-driven fiber maturation. A better understanding of how mechanical cues regulate fiber formation will help to develop better engineered replacements and rehabilitation protocols to drive repair after injury.
Collapse
Affiliation(s)
- Leia D Troop
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA 23284, United States.
| |
Collapse
|
2
|
Su X, Wang Y, Chen J, Liang Z, Wan L, Tang G. A feasibility study of in vivo quantitative ultra-short echo time-MRI for detecting early cartilage degeneration. Insights Imaging 2024; 15:162. [PMID: 38922455 PMCID: PMC11208376 DOI: 10.1186/s13244-024-01734-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVES To explore the feasibility of Ultra-short echo time (UTE) - MRI quantitative imaging in detecting early cartilage degeneration in vivo and underlying pathological and biochemical basis. METHODS Twenty volunteers with osteoarthritis (OA) planning for total knee arthroplasty (TKA) were prospectively recruited. UTE-MRI sequences and conventional sequences were performed preoperatively. Regions of interests (ROIs) were manually drawn on the tibial plateau and lateral femoral condyle images to calculate MRI values. Cartilage samples were collected during TKA according to the preset positions corresponding to MR images. Pathological and biochemical components of the corresponding ROI, including histological grading, glycosaminoglycan (GAG) content, collagen integrity, and water content were obtained. RESULTS 91 ROIs from volunteers of 7 males (age range: 68 to 78 years; 74 ± 3 years) and 13 females (age range: 57 to 79 years; 67 ± 6 years) were evaluated. UTE-MTR (r = -0.619, p < 0.001), UTE-AdiabT1ρ (r = 0.568, p < 0.001), and UTE-T2* values (r = -0.495, p < 0.001) showed higher correlation with Mankin scores than T2 (r = 0.287, p = 0.006) and T1ρ (r = 0.435, p < 0.001) values. Of them, UTE-MTR had the highest diagnostic performance (AUC = 0.824, p < 0.001). UTE-MTR, UTE-AdiabT1ρ and UTE-T2* value was mainly related to collagen structural integrity, PG content and water content, respectively (r = 0.536, -0.652, -0.518, p < 0.001, respectively). CONCLUSION UTE-MRI have shown greater in vivo diagnostic value for early cartilage degeneration compared to conventional T2 and T1ρ values. Of them, UTE-MTR has the highest diagnostic efficiency. UTE-MTR, UTE-AdiabT1ρ, and UTE-T2* value mainly reflect different aspects of cartilage degeneration--integrity of collagen structure, PG content, and water content, respectively. CRITICAL RELEVANCE STATEMENT Ultra-short echo time (UTE)-MRI has the potential to be a novel image biomarkers for detecting early cartilage degeneration in vivo and was correlated with biochemical changes of early cartilage degeneration. KEY POINTS Conventional MR may miss some early cartilage changes due to relatively long echo times. Ultra-short echo time (UTE)-MRI showed the ability in identifying early cartilage degeneration in vivo. UTE-MT, UTE-AdiabT1ρ, and UTE-T2* mapping mainly reflect different aspects of cartilage degeneration.
Collapse
Affiliation(s)
- Xiaolian Su
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yitong Wang
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jieying Chen
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zonghui Liang
- Department of Radiology, Shanghai Jing'an District Central Hospital, Shanghai, China
| | - Lidi Wan
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Chongming Branch of Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.
| | - Guangyu Tang
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Department of Radiology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Vernice NA, Dong X, Matavosian AA, Corpuz GS, Shin J, Bonassar LJ, Spector JA. Bioengineering Full-scale auricles using 3D-printed external scaffolds and decellularized cartilage xenograft. Acta Biomater 2024; 179:121-129. [PMID: 38494083 DOI: 10.1016/j.actbio.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/10/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Reconstruction of the human auricle remains a formidable challenge for plastic surgeons. Autologous costal cartilage grafts and alloplastic implants are technically challenging, and aesthetic and/or tactile outcomes are frequently suboptimal. Using a small animal "bioreactor", we have bioengineered full-scale ears utilizing decellularized cartilage xenograft placed within a 3D-printed external auricular scaffold that mimics the size, shape, and biomechanical properties of the native human auricle. The full-scale polylactic acid ear scaffolds were 3D-printed based upon data acquired from 3D photogrammetry of an adult ear. Ovine costal cartilage was processed either through mincing (1 mm3) or zesting (< 0.5 mm3), and then fully decellularized and sterilized. At explantation, both the minced and zested neoears maintained the size and contour complexities of the scaffold topography with steady tissue ingrowth through 6 months in vivo. A mild inflammatory infiltrate at 3 months was replaced by homogenous fibrovascular tissue ingrowth enveloping individual cartilage pieces at 6 months. All ear constructs were pliable, and the elasticity was confirmed by biomechanical analysis. Longer-term studies of the neoears with faster degrading biomaterials will be warranted for future clinical application. STATEMENT OF SIGNIFICANCE: Accurate reconstruction of the human auricle has always been a formidable challenge to plastic surgeons. In this article, we have bioengineered full-scale ears utilizing decellularized cartilage xenograft placed within a 3D-printed external auricular scaffold that mimic the size, shape, and biomechanical properties of the native human auricle. Longer-term studies of the neoears with faster degrading biomaterials will be warranted for future clinical application.
Collapse
Affiliation(s)
- Nicholas A Vernice
- Laboratory of Bioregenerative Medicine & Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Xue Dong
- Laboratory of Bioregenerative Medicine & Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Alicia A Matavosian
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - George S Corpuz
- Laboratory of Bioregenerative Medicine & Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, USA
| | - James Shin
- Department of Radiology, Well Cornell Medicine, New York, NY, USA
| | - Lawrence J Bonassar
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Jason A Spector
- Laboratory of Bioregenerative Medicine & Surgery, Department of Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, USA; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Troop LD, Puetzer JL. Intermittent Cyclic Stretch of Engineered Ligaments Drives Hierarchical Collagen Fiber Maturation in a Dose- and Organizational-Dependent Manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588420. [PMID: 38645097 PMCID: PMC11030411 DOI: 10.1101/2024.04.06.588420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Hierarchical collagen fibers are the primary source of strength in tendons and ligaments, however these fibers do not regenerate after injury or with repair, resulting in limited treatment options. We previously developed a culture system that guides ACL fibroblasts to produce native-sized fibers and fascicles by 6 weeks. These constructs are promising ligament replacements, but further maturation is needed. Mechanical cues are critical for development in vivo and in engineered tissues; however, the effect on larger fiber and fascicle formation is largely unknown. Our objective was to investigate whether intermittent cyclic stretch, mimicking rapid muscle activity, drives further maturation in our system to create stronger engineered replacements and to explore whether cyclic loading has differential effects on cells at different degrees of collagen organization to better inform engineered tissue maturation protocols. Constructs were loaded with an established intermittent cyclic loading regime at 5 or 10% strain for up to 6 weeks and compared to static controls. Cyclic loading drove cells to increase hierarchical collagen organization, collagen crimp, and tissue mechanics, ultimately producing constructs that matched or exceeded immature ACL properties. Further, the effect of loading on cells varied depending on degree of organization. Specifically, 10% load drove early improvements in mechanics and composition, while 5% load was more beneficial later in culture, suggesting a cellular threshold response and a shift in mechanotransduction. This study provides new insight into how cyclic loading affects cell-driven hierarchical fiber formation and maturation, which will help to develop better rehabilitation protocols and engineer stronger replacements.
Collapse
Affiliation(s)
- Leia D. Troop
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Jennifer L. Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States
| |
Collapse
|
5
|
Jenkins TL, Sarmiento Huertas PA, Umemori K, Guilak F, Little D. Tendon-derived matrix crosslinking techniques for electrospun multi-layered scaffolds. J Biomed Mater Res A 2023; 111:1875-1887. [PMID: 37489733 PMCID: PMC10592356 DOI: 10.1002/jbm.a.37588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023]
Abstract
Tendon tears are common and healing often occurs incompletely and by fibrosis. Tissue engineering seeks to improve repair, and one approach under investigation uses cell-seeded scaffolds containing biomimetic factors. Retention of biomimetic factors on the scaffolds is likely critical to maximize their benefit, while minimizing the risk of adverse effects, and without losing the beneficial effects of the biomimetic factors. The aim of the current study was to evaluate cross-linking methods to enhance the retention of tendon-derived matrix (TDM) on electrospun poly(ε-caprolactone) (PCL) scaffolds. We tested the effects of ultraviolet (UV) or carbodiimide (EDC:NHS:COOH) crosslinking methods to better retain TDM to the scaffolds and stimulate tendon-like matrix synthesis. Initially, we tested various crosslinking configurations of carbodiimide (2.5:1:1, 5:2:1, and 10:4:1 EDC:NHS:COOH ratios) and UV (30 s 1 J/cm2 , 60 s 1 J/cm2 , and 60 s 4 J/cm2 ) on PCL films compared to un-crosslinked TDM. We found that no crosslinking tested retained more TDM than coating alone (Kruskal-Wallis: p > .05), but that human adipose stem cells (hASCs) spread most on the 60 s 1 J/cm2 UV- and 2.5:1:1 EDC-crosslinked films (Kruskal-Wallis: p < .05). Next, we compared the effects of 60 s 1 J/cm2 UV- and 2.5:1:1 EDC-crosslinked to TDM-coated and untreated PCL scaffolds on hASC-induced tendon-like differentiation. UV-crosslinked scaffolds had greater modulus and stiffness than PCL or TDM scaffolds, and hASCs spread more on UV-crosslinked scaffolds (ANOVA: p < .05). Fourier transform infrared spectra revealed that UV- or EDC-crosslinking TDM did not affect the peaks at wavenumbers characteristic of tendon. Crosslinking TDM to electrospun scaffolds improves tendon-like matrix synthesis, providing a viable strategy for improving retention of TDM on electrospun PCL scaffolds.
Collapse
Affiliation(s)
- Thomas L. Jenkins
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | | | - Kentaro Umemori
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO
- Shriners Hospitals for Children – St. Louis, St. Louis, MO
| | - Dianne Little
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN
| |
Collapse
|
6
|
Brown ME, Puetzer JL. Enthesis maturation in engineered ligaments is differentially driven by loads that mimic slow growth elongation and rapid cyclic muscle movement. Acta Biomater 2023; 172:106-122. [PMID: 37839633 DOI: 10.1016/j.actbio.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
Entheses are complex attachments that translate load between elastic-ligaments and stiff-bone via organizational and compositional gradients. Neither natural healing, repair, nor engineered replacements restore these gradients, contributing to high re-tear rates. Previously, we developed a culture system which guides ligament fibroblasts in high-density collagen gels to develop early postnatal-like entheses, however further maturation is needed. Mechanical cues, including slow growth elongation and cyclic muscle activity, are critical to enthesis development in vivo but these cues have not been widely explored in engineered entheses and their individual contribution to maturation is largely unknown. Our objective here was to investigate how slow stretch, mimicking ACL growth rates, and intermittent cyclic loading, mimicking muscle activity, individually drive enthesis maturation in our system so to shed light on the cues governing enthesis development, while further developing our tissue engineered replacements. Interestingly, we found these loads differentially drive organizational maturation, with slow stretch driving improvements in the interface/enthesis region, and cyclic load improving the ligament region. However, despite differentially affecting organization, both loads produced improvements to interface mechanics and zonal composition. This study provides insight into how mechanical cues differentially affect enthesis development, while producing some of the most organized engineered enthesis to date. STATEMENT OF SIGNIFICANCE: Entheses attach ligaments to bone and are critical to load transfer; however, entheses do not regenerate with repair or replacement, contributing to high re-tear rates. Mechanical cues are critical to enthesis development in vivo but their individual contribution to maturation is largely unknown and they have not been widely explored in engineered replacements. Here, using a novel culture system, we provide new insight into how slow stretch, mimicking ACL growth rates, and intermittent cyclic loading, mimicking muscle activity, differentially affect enthesis maturation in engineered ligament-to-bone tissues, ultimately producing some of the most organized entheses to date. This system is a promising platform to explore cues regulating enthesis formation so to produce functional engineered replacements and better drive regeneration following repair.
Collapse
Affiliation(s)
- M Ethan Brown
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States.
| |
Collapse
|
7
|
Slyker L, Bonassar LJ. Alginate Conjugation Increases Toughness in Auricular Chondrocyte Seeded Collagen Hydrogels. Bioengineering (Basel) 2023; 10:1037. [PMID: 37760139 PMCID: PMC10526064 DOI: 10.3390/bioengineering10091037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Current auricular cartilage replacements for pediatric microtia fail to address the need for long-term integration and neocartilage formation. While collagen hydrogels have been successful in fostering neocartilage formation, the toughness and extensibility of these materials do not match that of native tissue. This study used the N-terminal functionalization of collagen with alginate oligomers to improve toughness and extensibility through metal-ion complexation. Alginate conjugation was confirmed via FTIR spectroscopy. The retention of native collagen fibrillar structure, thermal gelation, and helical conformation in functionalized gels was confirmed via scanning electron microscopy, oscillatory shear rheology, and circular dichroism spectroscopy, respectively. Alginate-calcium complexation enabled a more than two-fold increase in modulus and work density in functionalized collagen with the addition of 50 mM CaCl2, whereas unmodified collagen decreased in both modulus and work density with increasing calcium concentration. Additionally, the extensibility of alginate-functionalized collagen was increased at 25 and 50 mM CaCl2. Following 2-week culture with auricular chondrocytes, alginate-functionalization had no effect on the cytocompatibility of collagen gels, with no effects on cell density, and increased glycosaminoglycan deposition. Custom MATLAB video analysis was then used to quantify fracture toughness, which was more than 5-fold higher following culture in functionalized collagen and almost three-fold higher in unmodified collagen.
Collapse
Affiliation(s)
- Leigh Slyker
- Meinig of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lawrence J. Bonassar
- Meinig of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
8
|
Bates ME, Troop L, Brown ME, Puetzer JL. Temporal application of lysyl oxidase during hierarchical collagen fiber formation differentially effects tissue mechanics. Acta Biomater 2023; 160:98-111. [PMID: 36822485 PMCID: PMC10064799 DOI: 10.1016/j.actbio.2023.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
The primary source of strength in menisci, tendons, and ligaments are hierarchical collagen fibers; however, these fibers are not regenerated after injury nor in engineered replacements, resulting in limited repair options. Collagen strength is reliant on fiber alignment, density, diameter, and crosslinking. Recently, we developed a culture system which guides cells in high-density collagen gels to develop native-like hierarchically organized collagen fibers, which match native fiber alignment and diameters by 6 weeks. However, tensile moduli plateau at 1MPa, suggesting crosslinking may be lacking. Collagen crosslinking is regulated by lysyl oxidase (LOX) which forms immature crosslinks that condense into mature trivalent crosslinks. Trivalent crosslinks are thought to be the primarily source of strength in fibers, but it's not well understood how they form. The objective of this study was to evaluate the effect of exogenous LOX in our culture system at different stages of hierarchical fiber formation to produce stronger replacements and to better understand factors affecting crosslink maturation. We found treatment with LOX isoform LOXL2 did not restrict hierarchical fiber formation, with constructs still forming aligned collagen fibrils by 2 weeks, larger fibers by 4 weeks, and early fascicles by 6 weeks. However, LOXL2 treatment did significantly increase mature pyridinium (PYD) crosslink accumulation and tissue mechanics, with timing of LOXL2 supplementation during fiber formation having a significant effect. Overall, we found one week of LOXL2 supplementation at 4 weeks produced constructs with native-like fiber organization, increased PYD accumulation, and increased mechanics, ultimately matching the tensile modulus of immature bovine menisci. STATEMENT OF SIGNIFICANCE: Collagen fibers are the primary source of strength and function in connective tissues throughout the body, however it remains a challenge to develop these fibers in engineered replacements, greatly reducing treatment options. Here we demonstrate lysyl oxidase like 2 (LOXL2) can be used to significantly improve the mechanics of tissue engineered constructs, but timing of application is important and will most likely depend on degree of collagen organization or maturation. Currently there is limited understanding of how collagen crosslinking is regulated, and this system is a promising platform to further investigate cellular regulation of LOX crosslinking. Understanding the mechanism that regulates LOX production and activity is needed to ultimately regenerate functional repair or replacements for connective tissues throughout the body.
Collapse
Affiliation(s)
- Madison E Bates
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - Leia Troop
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - M Ethan Brown
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States.
| |
Collapse
|
9
|
Kim J, Bonassar LJ. Controlling collagen gelation pH to enhance biochemical, structural, and biomechanical properties of tissue-engineered menisci. J Biomed Mater Res A 2023; 111:478-487. [PMID: 36300869 DOI: 10.1002/jbm.a.37464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/16/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
Collagen-based hydrogels have been widely used in biomedical applications due to their biocompatibility. Enhancing mechanical properties of collagen gels remains challenging while maintaining biocompatibility. Here, we demonstrate that gelation pH has profound effects on cellular activity, collagen fibril structure, and mechanical properties of the fibrochondrocyte-seeded collagen gels in both short- and long-terms. Acidic and basic gelation pH, below pH 7.0 and above 8.5, resulted in dramatic cell death. Gelation pH ranging from 7.0 to 8.5 showed a relatively high cell viability. Furthermore, physiologic gelation (pH 7.5) showed the greatest collagen deposition while glycosaminoglycan deposition appeared independent of gelation pH. Scanning electron microscopy showed that neutral and physiologic gelation pH, 7.0 and 7.5, exhibited well-aligned collagen fibril structure on day 0 and enhanced collagen fibril structure with laterally joined fibrils on day 30. However, basic pH, 8.0 and 8.5, displayed a densely packed collagen fibril structure on day 0, which was also persistent on day 30. Initial equilibrium modulus increased with increasing gelation pH. Notably, after 30 days of culture, gelation pH of 7.5 and 8.0 showed the highest equilibrium modulus, reaching 150 -160 kPa. While controlling gelation pH is simply achieved compared with other strategies to improve mechanical properties, its influences on biochemical and biomechanical properties of the collagen gel are long-lasting. As such, gelation pH is a useful means to modulate both biochemical and biomechanical properties of the collagen-based hydrogels and can be utilized for diverse types of tissue engineering due to its simple application.
Collapse
Affiliation(s)
- Jongkil Kim
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
10
|
Lopez SG, Kim J, Estroff LA, Bonassar LJ. Removal of GAGs Regulates Mechanical Properties, Collagen Fiber Formation, and Alignment in Tissue Engineered Meniscus. ACS Biomater Sci Eng 2023; 9:1608-1619. [PMID: 36802372 DOI: 10.1021/acsbiomaterials.3c00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
The complex fibrillar architecture of native meniscus is essential for proper function and difficult to recapitulate in vitro. In the native meniscus, proteoglycan content is low during the development of collagen fibers and progressively increases with aging. In vitro, fibrochondrocytes produce glycosaminoglycans (GAGs) early in culture, in contrast to native tissue, where they are deposited after collagen fibers have formed. This difference in the timing of GAG production hinders the formation of a mature fiber network in such in vitro models. In this study, we removed GAGs from collagen gel-based tissue engineered constructs using chondroitinase ABC (cABC) and evaluated the effect on the formation and alignment of collagen fibers and the subsequent effect on tensile and compressive mechanical properties. Removal of GAGs during maturation of in vitro constructs improved collagen fiber alignment in tissue engineered meniscus constructs. Additionally, removal of GAGs during maturation improved fiber alignment without compromising compressive strength, and this removal improved not only fiber alignment and formation but also tensile properties. The increased fiber organization in cABC-treated groups also appeared to influence the size, shape, and location of defects in these constructs, suggesting that treatment may prevent the propagation of large defects under loading. This data gives another method of modulating the ECM for improved collagen fiber formation and mechanical properties in tissue engineered constructs.
Collapse
Affiliation(s)
- Serafina G Lopez
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Jongkil Kim
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States
- Kavli Institute for Nanoscale Science at Cornell, Cornell University, Ithaca, New York 14853, United States
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
11
|
Torzilli PA, Allen SN. Effect of Articular Surface Compression on Cartilage Extracellular Matrix Deformation. J Biomech Eng 2022; 144:091007. [PMID: 35292801 PMCID: PMC10782873 DOI: 10.1115/1.4054108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/28/2022] [Indexed: 11/08/2022]
Abstract
Early stage osteoarthritis is characterized by disruption of the superficial zone (SZ) of articular cartilage, including collagen damage and proteoglycan loss, resulting in "mechanical softening" of the extracellular matrix (ECM). The role of the SZ in controlling fluid exudation and imbibition during loading and unloading, respectively, was studied using confined creep compression tests. Bovine osteochondral (OC) plugs were subjected to either a static (88 kPa) or cyclic (0-125 kPa at 1 Hz) compressive stress for five minutes, and the cartilage deformation and recovery were measured during tissue loading and unloading, respectively. During unloading, the articular surface of the cartilage was either loaded with a small 1% tare load (∼1 kPa) applied through a porous load platen (covered), or completely unloaded (uncovered). Then the SZ (∼10%) of the cartilage was removed and the creep tests were repeated. Randomized tests were performed on each OC specimen to assess variability within and between plugs. Static creep strain was always greater than cyclic creep strain except at the beginning of loading (10-20 cycles). Uncovering the articular surface after creep deformation resulted in faster thickness recovery compared to the covered recovery. Removal of the SZ resulted in increased static and cyclic creep strains, as well as an increase in the cyclic peak-to-peak strain envelope. Our results indicate that an intact SZ is essential for normal cartilage mechanical function during joint motion by controlling fluid exudation and imbibition, and concomitantly ECM deformation and recovery, when loaded and unloaded, respectively.
Collapse
Affiliation(s)
- Peter A. Torzilli
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, Research Division 535, East 70th Street, New York, NY 10021
| | - Samie N. Allen
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY 10021
| |
Collapse
|
12
|
Gouldin AG, Brown ME, Puetzer JL. An inducible model for unraveling the effects of advanced glycation end-product accumulation in aging connective tissues. Connect Tissue Res 2022; 63:406-424. [PMID: 34706612 DOI: 10.1080/03008207.2021.1991333] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE In connective tissues there is a clear link between increasing age and degeneration. Advanced glycation end-products (AGEs) are believed to play a central role. AGEs are sugar-induced non-enzymatic crosslinks which accumulate in collagen with age and diabetes, altering tissue mechanics and cellular function. Despite ample correlative evidence linking collagen glycation to tissue degeneration, little is known how AGEs impact cell-matrix interactions, limiting therapeutic options. One reason for this limited understanding is that AGEs are typically induced using high concentrations of ribose which decrease cell viability, making it impossible to investigate cell-matrix interactions. The objective of this study was to develop a system to trigger AGE accumulation while maintaining cell viability. MATERIALS AND METHODS Using cell-seeded high density collagen gels, we investigated the effect of two systems for AGE induction, ribose at low concentrations (30, 100, and 200 mM) over 15 days of culture and riboflavin (0.25 and 0.75 mM) induced with blue light for 40 seconds (riboflavin-465 nm). RESULTS We found ribose and riboflavin-465 nm treatment produces fluorescent AGE quantities which match and/or exceed human fluorescent AGE levels for various tissues, ages, and diseases, without affecting cell viability or metabolism. Interestingly, a 40 second treatment of riboflavin-465 nm produced similar levels of fluorescent AGEs as 3 days of 100 mM ribose treatment. CONCLUSIONS Riboflavin-465 nm is a promising method to trigger AGEs on demand in vivo or in vitro without impacting cell viability and offers potential for unraveling the mechanism of AGEs in age and diabetes related tissue damage.
Collapse
Affiliation(s)
- Austin G Gouldin
- Departments of Biomedical Engineering; Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia, United States
| | - M Ethan Brown
- Departments of Biomedical Engineering; Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Jennifer L Puetzer
- Departments of Biomedical Engineering; Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
13
|
Wan L, Cheng X, Searleman AC, Ma YJ, Wong JH, Meyer RS, Du J, Tang G, Chang EY. Evaluation of enzymatic proteoglycan loss and collagen degradation in human articular cartilage using ultrashort echo time-based biomarkers: A feasibility study. NMR IN BIOMEDICINE 2022; 35:e4664. [PMID: 34904305 PMCID: PMC9042587 DOI: 10.1002/nbm.4664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 05/02/2023]
Abstract
The objective of the current study was to investigate the feasibility of quantitative 3D ultrashort echo time (UTE)-based biomarkers in detecting proteoglycan (PG) loss and collagen degradation in human cartilage. A total of 104 cartilage samples were harvested for a trypsin digestion study (n = 44), and a sequential trypsin and collagenase digestion study (n = 60), respectively. Forty-four cartilage samples were randomly divided into a trypsin digestion group (tryp group) and a control group (phosphate-buffered saline [PBS] group) (n = 22 for each group) for the trypsin digestion experiment. The remaining 60 cartilage samples were divided equally into four groups (n = 15 for each group) for sequential trypsin and collagenase digestion, including PBS + Tris (incubated in PBS, then Tris buffer solution), PBS + 30 U col (incubated in PBS, then 30 U/ml collagenase [30 U col] with Tris buffer solution), tryp + 30 U col (incubated in trypsin solution, then 30 U/ml collagenase with Tris buffer solution), and tryp + Tris (incubated in trypsin solution, then Tris buffer solution). The 3D UTE-based MRI biomarkers included T1 , multiecho T2 *, adiabatic T1ρ (AdiabT1ρ ), magnetization transfer ratio (MTR), and modeling of macromolecular proton fraction (MMF). For each cartilage sample, UTE-based biomarkers (T1 , T2 *, AdiabT1ρ , MTR, and MMF) and sample weight were evaluated before and after treatment. PG and hydroxyproline assays were performed. Differences between groups and correlations were assessed. All the evaluated biomarkers were able to differentiate between healthy and degenerated cartilage in the trypsin digestion experiment, but only T1 and AdiabT1ρ were significantly correlated with the PG concentration in the digestion solution (p = 0.004 and p = 0.0001, respectively). In the sequential digestion experiment, no significant differences were found for T1 and AdiabT1ρ values between the PBS + Tris and PBS + 30 U col groups (p = 0.627 and p = 0.877, respectively), but T1 and AdiabT1ρ values increased significantly in the tryp + Tris (p = 0.031 and p = 0.024, respectively) and tryp + 30 U col groups (both p < 0.0001). Significant decreases in MMF and MTR were found in the tryp + 30 U col group compared with the PBS + Tris group (p = 0.002 and p = 0.001, respectively). It was concluded that AdiabT1ρ and T1 have the potential for detecting PG loss, while MMF and MTR are promising for the detection of collagen degradation in articular cartilage, which could facilitate earlier, noninvasive diagnosis of osteoarthritis.
Collapse
Affiliation(s)
- Lidi Wan
- Department of Radiology, University of California, San Diego, CA
- Department of Radiology, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Xin Cheng
- Department of Radiology, University of California, San Diego, CA
- Division of Histology and Embryology, Jinan University, Guangzhou, China
| | | | - Ya-Jun Ma
- Department of Radiology, University of California, San Diego, CA
| | - Jonathan H. Wong
- Department of Radiology, University of California, San Diego, CA
- Radiology Service, VA San Diego Healthcare System, San Diego, CA
| | - R. Scott Meyer
- Orthopaedic Surgery Service, VA San Diego Healthcare System, San Diego, CA
| | - Jiang Du
- Department of Radiology, University of California, San Diego, CA
| | - Guangyu Tang
- Department of Radiology, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Eric Y. Chang
- Department of Radiology, University of California, San Diego, CA
- Radiology Service, VA San Diego Healthcare System, San Diego, CA
| |
Collapse
|
14
|
Yamaguchi J, Onodera T, Homan K, Liang X, Matsuoka M, Miyazaki T, Yoshiaki H, Saito M, Iwasaki N. Optical coherence tomography evaluation of the spatiotemporal effects of 3D bone marrow stromal cell culture using a bioreactor. J Biomed Mater Res B Appl Biomater 2022; 110:1853-1861. [PMID: 35262287 DOI: 10.1002/jbm.b.35043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 01/04/2022] [Accepted: 02/13/2022] [Indexed: 11/11/2022]
Abstract
Performing cell culture in a three-dimensional (3D) environment has various advantages. In cartilage tissue engineering, 3D in vitro cultures utilizing biomaterials and bioreactors can mimic the biological environment. However, the biggest drawback of these 3D culture systems is a limited ability to evaluate 3D cell distribution. Optical coherence tomography (OCT) has recently been used to evaluate 3D cellular morphology and structure in a timely manner. Here, we showed that OCT could be used to visually assess the distribution and the morphology of bone marrow stromal cells under chondrogenic 3D cultivation using alginate gels and rotary culture. In particular, OCT was able to visualize living cells embedded in alginate gels in a non-destructive and 3D manner, as well as quantitatively evaluate cell distribution and spheroid volume. We also found that cells were centralized in rotary culture but peripherally distributed in static culture, while rotary culture enhanced the hypertrophy of marrow stromal cells (MSCs) embedded in alginate gels. Together, our findings demonstrate that OCT can be used to evaluate the spatiotemporal effects of 3D cultivation using alginate gels and rotary culture. Therefore, this method may allow the observation of pre-cultured tissue over time and the optimization of culture conditions for regenerative tissue engineering.
Collapse
Affiliation(s)
- Jun Yamaguchi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomohiro Onodera
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kentaro Homan
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Xu Liang
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masatake Matsuoka
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takuji Miyazaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hosokawa Yoshiaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
15
|
Lintz M, Walk RE, Tang SY, Bonassar LJ. The degenerative impact of hyperglycemia on the structure and mechanics of developing murine intervertebral discs. JOR Spine 2022; 5:e1191. [PMID: 35386755 PMCID: PMC8966876 DOI: 10.1002/jsp2.1191] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/30/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Diabetes has long been implicated as a major risk factor for intervertebral disc (IVD) degeneration, interfering with molecular signaling and matrix biochemistry, which ultimately aggravates the progression of the disease. Glucose content has been previously shown to influence structural and compositional changes in engineered discs in vitro, impeding fiber formation and mechanical stability. Methods In this study, we investigated the impact of diabetic hyperglycemia on young IVDs by assessing biochemical composition, collagen fiber architecture, and mechanical behavior of discs harvested from 3- to 4-month-old db/db mouse caudal spines. Results We found that discs taken from diabetic mice with elevated blood glucose levels demonstrated an increase in total glycosaminoglycan and collagen content, but comparable advanced glycation end products (AGE) levels to wild-type discs. Diabetic discs also contained ill-defined boundaries between the nucleus pulposus and annulus fibrosus, with the latter showing a disorganized and unaligned collagen fiber network at this same boundary. Conclusions These compositional and structural changes had a detrimental effect on function, as the diabetic discs were twice as stiff as their wild-type counterparts and demonstrated a significant resistance to deformation. These results indicate that diabetes may predispose the young disc to DDD later in life by altering patterns of extracellular matrix deposition, fiber formation, and motion segment mechanics independently of AGE accumulation.
Collapse
Affiliation(s)
- Marianne Lintz
- Meinig School of Biomedical EngineeringCornell UniversityIthacaNew YorkUSA
| | - Remy E. Walk
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Simon Y. Tang
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
- Department of Mechanical Engineering and Materials ScienceWashington University in St. LouisSt. LouisMissouriUSA
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Lawrence J. Bonassar
- Meinig School of Biomedical EngineeringCornell UniversityIthacaNew YorkUSA
- Sibley School of Mechanical and Aerospace EngineeringCornell UniversityIthacaNew YorkUSA
| |
Collapse
|
16
|
Driving native-like zonal enthesis formation in engineered ligaments using mechanical boundary conditions and β-tricalcium phosphate. Acta Biomater 2022; 140:700-716. [PMID: 34954418 DOI: 10.1016/j.actbio.2021.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 11/21/2022]
Abstract
Fibrocartilaginous entheses are structurally complex tissues that translate load from elastic ligaments to stiff bone via complex zonal gradients in the organization, mineralization, and cell phenotype. Currently, these complex gradients necessary for long-term mechanical function are not recreated in soft tissue-to-bone healing or engineered replacements, contributing to high failure rates. Previously, we developed a culture system that guides ligament fibroblasts to develop aligned native-sized collagen fibers using high-density collagen gels and mechanical boundary conditions. These constructs are promising ligament replacements, however functional ligament-to-bone attachments, or entheses, are required for long-term function in vivo. The objective of this study was to investigate the effect of compressive mechanical boundary conditions and the addition of beta-tricalcium phosphate (βTCP), a known osteoconductive agent, on the development of zonal ligament-to-bone entheses. We found that compressive boundary clamps, that restrict cellular contraction and produce a zonal tensile-compressive environment, guide ligament fibroblasts to produce 3 unique zones of collagen organization and zonal accumulation of glycosaminoglycans (GAGs), type II, and type X collagen. Ultimately, by 6 weeks of culture these constructs had similar organization and composition as immature bovine entheses. Further, βTCP applied under the clamp enhanced maturation of these entheses, leading to significantly increased tensile moduli, and zonal GAG accumulation, ALP activity, and calcium-phosphate accumulation, suggesting the initiation of endochondral ossification. This culture system produced some of the most organized entheses to date, closely mirroring early postnatal enthesis development, and provides an in vitro platform to better understand the cues that drive enthesis maturation in vivo. STATEMENT OF SIGNIFICANCE: Ligaments are attached to bone via entheses. Entheses are complex tissues with gradients in organization, composition, and cell phenotype. Entheses are necessary for proper transfer of load from ligament-to-bone, but currently are not restored with healing or replacements. Here, we provide new insight into how tensile-compressive boundary conditions and βTCP drive zonal gradients in collagen organization, mineralization, and matrix composition, producing tissues similar to immature ligament-to-bone attachments. Collectively, this culture system uses a bottom-up approach with mechanical and biochemical cues to produce engineered replacements which closely mirror postnatal enthesis development. This culture system is a promising platform to better understanding the cues that regulate enthesis formation so to better drive enthesis regeneration following graft repair and in engineered replacements.
Collapse
|
17
|
Sonthithai P, Hankamonsiri W, Lertwimol T, Uppanan P, Janvikul W. Novel modified culture medium for enhancing redifferentiation of chondrocytes for cartilage tissue engineering applications. Biotechnol Prog 2022; 38:e3240. [DOI: 10.1002/btpr.3240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Pacharapan Sonthithai
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park Phahonyothin Road, Klong Luang, Pathum Thani 12120 Thailand
| | - Weerawan Hankamonsiri
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park Phahonyothin Road, Klong Luang, Pathum Thani 12120 Thailand
| | - Tareerat Lertwimol
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park Phahonyothin Road, Klong Luang, Pathum Thani 12120 Thailand
| | - Paweena Uppanan
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park Phahonyothin Road, Klong Luang, Pathum Thani 12120 Thailand
| | - Wanida Janvikul
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park Phahonyothin Road, Klong Luang, Pathum Thani 12120 Thailand
| |
Collapse
|
18
|
Marshall SL, Jacobsen TD, Emsbo E, Murali A, Anton K, Liu JZ, Lu HH, Chahine NO. Three-Dimensional-Printed Flexible Scaffolds Have Tunable Biomimetic Mechanical Properties for Intervertebral Disc Tissue Engineering. ACS Biomater Sci Eng 2021; 7:5836-5849. [PMID: 34843224 DOI: 10.1021/acsbiomaterials.1c01326] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The intervertebral disc (IVD) exhibits complex structure and biomechanical function, which supports the weight of the body and permits motion. Surgical treatments for IVD degeneration (e.g., lumbar fusion, disc replacement) often disrupt the mechanical environment of the spine which lead to adjacent segment disease. Alternatively, disc tissue engineering strategies, where cell-seeded hydrogels or fibrous biomaterials are cultured in vitro to promote matrix deposition, do not recapitulate the complex IVD mechanical properties. In this study, we use 3D printing of flexible polylactic acid (FPLA) to fabricate a viscoelastic scaffold with tunable biomimetic mechanics for whole spine motion segment applications. We optimized the mechanical properties of the scaffolds for equilibrium and dynamic moduli in compression and tension by varying fiber spacing or porosity, generating scaffolds with de novo mechanical properties within the physiological range of spine motion segments. The biodegradation analysis of the 3D printed scaffolds showed that FPLA exhibits lower degradation rate and thus has longer mechanical stability than standard PLA. FPLA scaffolds were biocompatible, supporting viability of nucleus pulposus (NP) cells in 2D and in FPLA+hydrogel composites. Composite scaffolds cultured with NP cells maintained baseline physiological mechanical properties and promoted matrix deposition up to 8 weeks in culture. Mesenchymal stromal cells (MSCs) cultured on FPLA adhered to the scaffold and exhibited fibrocartilaginous differentiation. These results demonstrate for the first time that 3D printed FPLA scaffolds have de novo viscoelastic mechanical properties that match the native IVD motion segment in both tension and compression and have the potential to be used as a mechanically stable and biocompatible biomaterial for engineered disc replacement.
Collapse
Affiliation(s)
- Samantha L Marshall
- Department of Orthopedic Surgery, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Timothy D Jacobsen
- Department of Orthopedic Surgery, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States.,Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Erik Emsbo
- Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Archana Murali
- Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Kevin Anton
- Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Jessica Z Liu
- Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Helen H Lu
- Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| | - Nadeen O Chahine
- Department of Orthopedic Surgery, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States.,Department of Biomedical Engineering, Columbia University, 650 West 168th Street, 1410, New York, New York 10031, United States
| |
Collapse
|
19
|
Middendorf JM, Diamantides N, Kim B, Dugopolski C, Kennedy S, Blahut E, Cohen I, Bonassar LJ. The influence of chondrocyte source on the manufacturing reproducibility of human tissue engineered cartilage. Acta Biomater 2021; 131:276-285. [PMID: 34245892 DOI: 10.1016/j.actbio.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/16/2022]
Abstract
Multiple human tissue engineered cartilage constructs are showing promise in advanced clinical trials but identifying important measures of manufacturing reproducibility remains a challenge. FDA guidance suggests measuring multiple mechanical properties prior to implantation, because these properties could affect the long term success of the implant. Additionally, these engineered cartilage mechanics could be sensitive to the autologous chondrocyte source, an inherently irregular manufacturing starting material. If any mechanical properties are sensitive to changes in the autologous chondrocyte source, these properties may need to be measured prior to implantation to ensure manufacturing reproducibility and quality. Therefore, this study identified variability in the compressive, friction, and shear properties of a human tissue engineered cartilage constructs due to the chondrocyte source. Over 200 constructs were created from 7 different chondrocyte sources and tested using 3 distinct mechanical experiments. Under confined compression, the compressive properties (aggregate modulus and hydraulic permeability) varied by orders of magnitude due to the chondrocyte source. The friction coefficient changed by a factor of 5 due to the chondrocyte source and high intrapatient variability was noted. In contrast, the shear modulus was not affected by changes in the chondrocyte source. Finally, measurements on the local compressive and shear mechanics revealed variability in the depth dependent strain fields based on chondrocyte source. Since the chondrocyte source causes large amounts of variability in the compression and local mechanical properties of engineered cartilage, these mechanical properties may be important measures of manufacturing reproducibility. STATEMENT OF SIGNIFICANCE: Although the FDA recommends measuring mechanical properties of human tissue engineered cartilage constructs during manufacturing, the effect of manufacturing variability on construct mechanics is unknown. As one of the first studies to measure multiple mechanical properties on hundreds of human tissue engineered cartilage constructs, we found the compressive properties are most sensitive to changes in the autologous chondrocyte source, an inherently irregular manufacturing variable. This sensitivity to the autologous chondrocyte source reveals the compressive properties should be measured prior to implantation to assess manufacturing reproducibility.
Collapse
Affiliation(s)
- Jill M Middendorf
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, NY, United States
| | - Nicole Diamantides
- Meinig School of Biomedical Engineering Cornell University, Ithaca, NY, United States
| | - Byumsu Kim
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, NY, United States
| | | | | | - Eric Blahut
- Histogenics Corporation, Waltham, MA, United States
| | - Itai Cohen
- Department of Physics, Cornell University, Ithaca, NY, United States
| | - Lawrence J Bonassar
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, NY, United States; Meinig School of Biomedical Engineering Cornell University, Ithaca, NY, United States.
| |
Collapse
|
20
|
Mosher CZ, Brudnicki PAP, Gong Z, Childs HR, Lee SW, Antrobus RM, Fang EC, Schiros TN, Lu HH. Green electrospinning for biomaterials and biofabrication. Biofabrication 2021; 13. [PMID: 34102612 DOI: 10.1088/1758-5090/ac0964] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/08/2021] [Indexed: 11/12/2022]
Abstract
Green manufacturing has emerged across industries, propelled by a growing awareness of the negative environmental and health impacts associated with traditional practices. In the biomaterials industry, electrospinning is a ubiquitous fabrication method for producing nano- to micro-scale fibrous meshes that resemble native tissues, but this process traditionally utilizes solvents that are environmentally hazardous and pose a significant barrier to industrial scale-up and clinical translation. Applying sustainability principles to biomaterial production, we have developed a 'green electrospinning' process by systematically testing biologically benign solvents (U.S. Food and Drug Administration Q3C Class 3), and have identified acetic acid as a green solvent that exhibits low ecological impact (global warming potential (GWP) = 1.40 CO2eq. kg/L) and supports a stable electrospinning jet under routine fabrication conditions. By tuning electrospinning parameters, such as needle-plate distance and flow rate, we updated the fabrication of widely utilized biomedical polymers (e.g. poly-α-hydroxyesters, collagen), polymer blends, polymer-ceramic composites, and growth factor delivery systems. Resulting 'green' fibers and composites are comparable to traditional meshes in terms of composition, chemistry, architecture, mechanical properties, and biocompatibility. Interestingly, material properties of green synthetic fibers are more biomimetic than those of traditionally electrospun fibers, doubling in ductility (91.86 ± 35.65 vs. 45 ± 15.07%,n= 10,p< 0.05) without compromising yield strength (1.32 ± 0.26 vs. 1.38 ± 0.32 MPa) or ultimate tensile strength (2.49 ± 0.55 vs. 2.36 ± 0.45 MPa). Most importantly, green electrospinning proves advantageous for biofabrication, rendering a greater protection of growth factors during fiber formation (72.30 ± 1.94 vs. 62.87 ± 2.49% alpha helical content,n= 3,p< 0.05) and recapitulating native ECM mechanics in the fabrication of biopolymer-based meshes (16.57 ± 3.92% ductility, 33.38 ± 30.26 MPa elastic modulus, 1.30 ± 0.19 MPa yield strength, and 2.13 ± 0.36 MPa ultimate tensile strength,n= 10). The eco-conscious approach demonstrated here represents a paradigm shift in biofabrication, and will accelerate the translation of scalable biomaterials and biomimetic scaffolds for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Christopher Z Mosher
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Philip A P Brudnicki
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Zhengxiang Gong
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Hannah R Childs
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Sang Won Lee
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Romare M Antrobus
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Elisa C Fang
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Theanne N Schiros
- Materials Research Science and Engineering Center, Columbia University, New York, NY 10027, United States of America.,Science and Mathematics Department, Fashion Institute of Technology, New York, NY 10001, United States of America
| | - Helen H Lu
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America.,Materials Research Science and Engineering Center, Columbia University, New York, NY 10027, United States of America
| |
Collapse
|
21
|
Das P, Mishra R, Devi B, Rajesh K, Basak P, Roy M, Roy P, Lahiri D, Nandi SK. Decellularized xenogenic cartilage extracellular matrix (ECM) scaffolds for the reconstruction of osteochondral defects in rabbits. J Mater Chem B 2021; 9:4873-4894. [PMID: 34095925 DOI: 10.1039/d1tb00314c] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of decellularized native allogenic or xenogenic cartilaginous extracellular matrix (ECM) biomaterials is widely expanding in the fields of tissue engineering and regenerative medicine. In this study, we aimed to develop an acellular, affordable, biodegradable, easily available goat conchal cartilaginous ECM derived scaffolding biomaterial for repair and regeneration of osteochondral defects in rabbits. Cartilages harvested from freshly collected goat ears were decellularized using chemical agents, namely, hypotonic-hypertonic (HH) buffer and Triton X-100 solution, separately. The morphologies and ultrastructure orientations of the decellularized cartilages remained unaltered in spite of complete cellular loss. Furthermore, when the acellular cartilaginous ECMs were cultured with murine mesenchymal stem cells (MSCs) (C3H10T1/2 cells), cellular infiltration and proliferation were thoroughly monitored using SEM, DAPI and FDA stained images, whereas the MTT assay proved the biocompatibility of the matrices. The increasing amounts of secreted ECM proteins (collagen and sGAG) indicated successful chondrogenic differentiation of the MSCs in the presence of the treated cartilage samples. In vivo biocompatibility studies showed no significant immune response or tissue rejection in the treated samples but tissue necrosis in control samples after 3 months. Upon implantation of the constructs in rabbits' osteochondral defects for 3 months, the histological and micro-CT evaluation revealed significant enhancement and regeneration of neocartilage and subchondral bony tissues. The IGF-1 loaded cartilaginous constructs showed comparatively better healing response after 3 months. Our results showed that decellularized xenogenic cartilaginous biomaterials preserved the bioactivity and integrity of the matrices that also favored in vitro stem cell proliferation and chondrogenic differentiation and enabled osteochondral regeneration, thus paving a new way for articular cartilage reconstruction.
Collapse
Affiliation(s)
- Piyali Das
- School of Bioscience and Engineering, Jadavpur University, Kolkata, India
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kim J, Boys AJ, Estroff LA, Bonassar LJ. Combining TGF-β1 and Mechanical Anchoring to Enhance Collagen Fiber Formation and Alignment in Tissue-Engineered Menisci. ACS Biomater Sci Eng 2021; 7:1608-1620. [PMID: 33606521 DOI: 10.1021/acsbiomaterials.0c01791] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Recapitulating the collagen fiber structure of native menisci is one of the major challenges in the development of tissue-engineered menisci. Native collagen fibers are developed by the complex interplay of biochemical and biomechanical signals. In this study, we optimized glucose and transforming growth factor-β1 (TGF-β1) concentrations in combination with mechanical anchoring to balance contributions of proteoglycan synthesis and contractile behavior in collagen fiber assembly. Glucose had a profound effect on the final dimensions of collagen-based constructs. TGF-β1 influenced construct contraction rate and glycosaminoglycan (GAG) production with two half-maximal effective concentration (EC50) ranges, which are 0.23 to 0.28 and 0.53 to 1.71 ng/mL, respectively. At concentrations less than the EC50, for the GAG production and contraction rate, TGF-β1 treatment resulted in less organized collagen fibers. At concentrations greater than the EC50, TGF-β1 led to dense, disorganized collagen fibers. Between the two EC50 values, collagen fiber diameter and length increased. The effects of TGF-β1 on fiber development were enhanced by mechanical anchoring, leading to peaks in fiber diameter, length, and alignment index. Fiber diameter and length increased from 7.9 ± 1.4 and 148.7 ± 16.4 to 17.5 ± 2.1 and 262.0 ± 13.0 μm, respectively. The alignment index reached 1.31, comparable to that of native tissue, 1.40. These enhancements in fiber architecture resulted in significant increases in tensile modulus and ultimate tensile stress (UTS) by 1.6- and 1.4-fold. Correlation analysis showed that tensile modulus and UTS strongly correlated with collagen fiber length, diameter, and alignment, while compressive modulus correlated with GAG content. These outcomes highlight the need for optimization of both biochemical and biomechanical cues in the culture environment for enhancing fiber development within tissue-engineered constructs.
Collapse
Affiliation(s)
- Jongkil Kim
- Meinig of Biomedical Engineering, Cornell University, 237 Tower Road, Ithaca, New York 14853, United States
| | - Alexander J Boys
- Department of Materials Science and Engineering, Cornell University, 126 Hollister Drive, Ithaca, New York 14853, United States
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, 126 Hollister Drive, Ithaca, New York 14853, United States.,Kavli Institute at Cornell for Nanoscale Science, Cornell University, 245 East Avenue, Ithaca, New York 14853, United States
| | - Lawrence J Bonassar
- Meinig of Biomedical Engineering, Cornell University, 237 Tower Road, Ithaca, New York 14853, United States.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, 313 Campus Road, Ithaca, New York 14853, United States
| |
Collapse
|
23
|
Zhang P, Liu Y, Jia L, Ci Z, Zhang W, Liu Y, Chen J, Cao Y, Zhou G. SP600125, a JNK-Specific Inhibitor, Regulates in vitro Auricular Cartilage Regeneration by Promoting Cell Proliferation and Inhibiting Extracellular Matrix Metabolism. Front Cell Dev Biol 2021; 9:630678. [PMID: 33816478 PMCID: PMC8010669 DOI: 10.3389/fcell.2021.630678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/15/2021] [Indexed: 11/24/2022] Open
Abstract
In vitro construction is a major trend involved in cartilage regeneration and repair. Satisfactory in vitro cartilage regeneration depends on a suitable culture system. Current chondrogenic culture systems with a high content of transforming growth factor beta-1 effectively promote cartilaginous extracellular matrix (ECM) production but inhibit chondrocyte survival. As is known, inhibition of the c-Jun N-terminal kinase (JNK) signaling pathway acts in blocking the progression of osteoarthritis by reducing chondrocyte apoptosis and cartilage destruction. However, whether inhibiting JNK signaling resists the inhibitory effect of current chondrogenic medium (CM) on cell survival and affects in vitro auricular cartilage regeneration (including cell proliferation, ECM synthesis, and degradation) has not been investigated. In order to address these issues and optimize the chondrogenic culture system, we generated a three-dimensional in vitro auricular cartilage regeneration model to investigate the effects of SP600125 (a JNK-specific inhibitor) on chondrocyte proliferation and ECM metabolism. SP600125 supplementation efficiently promoted cell proliferation at both cellular and tissue levels and canceled the negative effect of our chondrogenic culture system on cell survival. Moreover, it significantly inhibited ECM degradation by reducing the expressions of tumor necrosis factor-alpha, interleukin-1-beta, and matrix metalloproteinase 13. In addition, SP600125 inhibited ECM synthesis at both cellular and tissue levels, but this could be canceled and even reversed by adding chondrogenic factors; yet this enabled a sufficient number of chondrocytes to be retained at the same time. Thus, SP600125 had a positive effect on in vitro auricular cartilage regeneration in terms of cell proliferation and ECM degradation but a negative effect on ECM synthesis, which could be reversed by adding CM. Therefore, a combination of SP600125 and CM might help in optimizing current chondrogenic culture systems and achieve satisfactory in vitro cartilage regeneration by promoting cell proliferation, reducing ECM degradation, and enhancing ECM synthesis.
Collapse
Affiliation(s)
- Peiling Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqun Liu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China
| | - Litao Jia
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China
| | - Zheng Ci
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China
| | - Wei Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China
| | - Yu Liu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Jie Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,National Tissue Engineering Center of China, Shanghai, China
| |
Collapse
|
24
|
Ci Z, Zhang Y, Wang Y, Wu G, Hou M, Zhang P, Jia L, Bai B, Cao Y, Liu Y, Zhou G. 3D Cartilage Regeneration With Certain Shape and Mechanical Strength Based on Engineered Cartilage Gel and Decalcified Bone Matrix. Front Cell Dev Biol 2021; 9:638115. [PMID: 33718376 PMCID: PMC7952450 DOI: 10.3389/fcell.2021.638115] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/26/2021] [Indexed: 01/09/2023] Open
Abstract
Scaffold-free cartilage-sheet technology can stably regenerate high-quality cartilage tissue in vivo. However, uncontrolled shape maintenance and mechanical strength greatly hinder its clinical translation. Decalcified bone matrix (DBM) has high porosity, a suitable pore structure, and good biocompatibility, as well as controlled shape and mechanical strength. In this study, cartilage sheet was prepared into engineered cartilage gel (ECG) and combined with DBM to explore the feasibility of regenerating 3D cartilage with controlled shape and mechanical strength. The results indicated that ECG cultured in vitro for 3 days (3 d) and 15 days (15 d) showed good biocompatibility with DBM, and the ECG–DBM constructs successfully regenerated viable 3D cartilage with typical mature cartilage features in both nude mice and autologous goats. Additionally, the regenerated cartilage had comparable mechanical properties to native cartilage and maintained its original shape. To further determine the optimal seeding parameters for ECG, the 3 d ECG regenerated using human chondrocytes was diluted in different concentrations (1:3, 1:2, and 1:1) for seeding and in vivo implantation. The results showed that the regenerated cartilage in the 1:2 group exhibited better shape maintenance and homogeneity than the other groups. The current study established a novel mode of 3D cartilage regeneration based on the design concept of steel (DBM)-reinforced concrete (ECG) and successfully regenerated homogenous and mature 3D cartilage with controlled shape and mechanical strength, which hopefully provides an ideal cartilage graft for the repair of various cartilage defects.
Collapse
Affiliation(s)
- Zheng Ci
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Ying Zhang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahui Wang
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Gaoyang Wu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Mengjie Hou
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Peiling Zhang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Litao Jia
- National Tissue Engineering Center of China, Shanghai, China.,Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoshuai Bai
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Yilin Cao
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Yu Liu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Guangdong Zhou
- Research Institute of Plastic Surgery, Wei Fang Medical College, Wei Fang, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| |
Collapse
|
25
|
Puetzer JL, Ma T, Sallent I, Gelmi A, Stevens MM. Driving Hierarchical Collagen Fiber Formation for Functional Tendon, Ligament, and Meniscus Replacement. Biomaterials 2021; 269:120527. [PMID: 33246739 PMCID: PMC7883218 DOI: 10.1016/j.biomaterials.2020.120527] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/09/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022]
Abstract
Hierarchical collagen fibers are the primary source of strength in musculoskeletal tendons, ligaments, and menisci. It has remained a challenge to develop these large fibers in engineered replacements or in vivo after injury. The objective of this study was to investigate the ability of restrained cell-seeded high density collagen gels to drive hierarchical fiber formation for multiple musculoskeletal tissues. We found boundary conditions applied to high density collagen gels were capable of driving tenocytes, ligament fibroblasts, and meniscal fibrochondrocytes to develop native-sized hierarchical collagen fibers 20-40 μm in diameter. The fibers organize similar to bovine juvenile collagen with native fibril banding patterns and hierarchical fiber bundles 50-350 μm in diameter by 6 weeks. Mirroring fiber organization, tensile properties of restrained samples improved significantly with time, reaching ~1 MPa. Additionally, tendon, ligament, and meniscal cells produced significantly different sized fibers, different degrees of crimp, and different GAG concentrations, which corresponded with respective juvenile tissue. To our knowledge, these are some of the largest, most organized fibers produced to date in vitro. Further, cells produced tissue specific hierarchical fibers, suggesting this system is a promising tool to better understand cellular regulation of fiber formation to better stimulate it in vivo after injury.
Collapse
Affiliation(s)
- Jennifer L Puetzer
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ; Department of Biomedical Engineering and Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States, 23284.
| | - Tianchi Ma
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Ignacio Sallent
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Amy Gelmi
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ.
| |
Collapse
|
26
|
Photocrosslinked natural hydrogel composed of hyaluronic acid and gelatin enhances cartilage regeneration of decellularized trachea matrix. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111628. [PMID: 33545814 DOI: 10.1016/j.msec.2020.111628] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Abstract
Repair of long segmental trachea defects is always a great challenge in the clinic. The key to solving this problem is to develop an ideal trachea substitute with biological function. Using of a decellularized trachea matrix based on laser micropore technique (LDTM) demonstrated the possibility of preparing ideal trachea substitutes with tubular shape and satisfactory cartilage regeneration for tissue-engineered trachea regeneration. However, as a result of the very low cell adhesion of LDTM, an overly high concentration of seeding cell is required, which greatly restricts its clinical translation. To address this issue, the current study proposed a novel strategy using a photocrosslinked natural hydrogel (PNH) carrier to enhance cell retention efficiency and improve tracheal cartilage regeneration. Our results demonstrated that PNH underwent a rapid liquid-solid phase conversion under ultraviolet light. Moreover, the photo-generated aldehyde groups in PNH could rapidly react with inherent amino groups on LDTM surfaces to form imine bonds, which efficiently immobilized the cell-PNH composite to the surfaces of LDTM and/or maintained the composite in the LDTM micropores. Therefore, PNH significantly enhanced cell-seeding efficiency and achieved both stable cell retention and homogenous cell distribution throughout the LDTM. Moreover, PNH exhibited excellent biocompatibility and low cytotoxicity, and provided a natural three-dimensional biomimetic microenvironment to efficiently promote chondrocyte survival and proliferation, extracellular matrix production, and cartilage regeneration. Most importantly, at a relatively low cell-seeding concentration, homogeneous tubular cartilage was successfully regenerated with an accurate tracheal shape, sufficient mechanical strength, good elasticity, typical lacuna structure, and cartilage-specific extracellular matrix deposition. Our findings establish a versatile and efficient cell-seeding strategy for regeneration of various tissue and provide a satisfactory trachea substitute for repair and functional reconstruction of long segmental tracheal defects.
Collapse
|
27
|
Neidlin M, Chantzi E, Macheras G, Gustafsson MG, Alexopoulos LG. A Novel Multiplex Based Platform for Osteoarthritis Drug Candidate Evaluation. Ann Biomed Eng 2020; 48:2438-2448. [PMID: 32472364 DOI: 10.1007/s10439-020-02539-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 01/10/2023]
Abstract
Osteoarthritis (OA) is characterized by irreversible cartilage degradation with very limited therapeutic interventions. Drug candidates targeted at prototypic players had limited success until now and systems based approaches might be necessary. Consequently, drug evaluation platforms should consider the biological complexity looking beyond well-known contributors of OA. In this study an ex vivo model of cartilage degradation, combined with measuring releases of 27 proteins, was utilized to study 9 drug candidates. After an initial single drug evaluation step the 3 most promising compounds were selected and employed in an exhaustive combinatorial experiment. The resulting most and least promising treatment candidates were selected and validated in an independent study. This included estimation of mechanical properties via finite element modelling (FEM) and quantification of cartilage degradation as glycosaminoglycan (GAG) release. The most promising candidate showed increase of Young's modulus, decrease of hydraulic permeability and decrease of GAG release. The least promising candidate exhibited the opposite behaviour. The study shows the potential of a novel drug evaluation platform in identifying treatments that might reduce cartilage degradation. It also demonstrates the promise of exhaustive combination experiments and a connection between chondrocyte responses at the molecular level with changes of biomechanical properties at the tissue level.
Collapse
Affiliation(s)
- Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, Heroon Polytechniou 9, 15780, Zografou, Greece
| | - Efthymia Chantzi
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Leonidas G Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, Heroon Polytechniou 9, 15780, Zografou, Greece.
| |
Collapse
|
28
|
Middendorf JM, Diamantides N, Shortkroff S, Dugopolski C, Kennedy S, Cohen I, Bonassar LJ. Multiscale mechanics of tissue-engineered cartilage grown from human chondrocytes and human-induced pluripotent stem cells. J Orthop Res 2020; 38:1965-1973. [PMID: 32125023 DOI: 10.1002/jor.24643] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 02/17/2020] [Accepted: 02/29/2020] [Indexed: 02/04/2023]
Abstract
Tissue-engineered cartilage has shown promising results in the repair of focal cartilage defects. However, current clinical techniques rely on an extra surgical procedure to biopsy healthy cartilage to obtain human chondrocytes. Alternatively, induced pluripotent stem cells (iPSCs) have the ability to differentiate into chondrocytes and produce cartilaginous matrix without the need to biopsy healthy cartilage. However, the mechanical properties of tissue-engineered cartilage with iPSCs are unknown and might be critical to long-term tissue function and health. This study used confined compression, cartilage on glass tribology, and shear testing on a confocal microscope to assess the macroscale and microscale mechanical properties of two constructs seeded with either chondrocyte-derived iPSCs (Ch-iPSCs) or native human chondrocytes. Macroscale properties of Ch-iPSC constructs provided similar or better mechanical properties than chondrocyte constructs. Under compression, Ch-iPSC constructs had an aggregate modulus that was two times larger than chondrocyte constructs and was closer to native tissue. No differences in the shear modulus and friction coefficients were observed between Ch-iPSC and chondrocyte constructs. On the microscale, Ch-iPSC and chondrocyte constructs had different depth-dependent mechanical properties, neither of which matches native tissue. These observed depth-dependent differences may be important to the function of the implant. Overall, this comparison of multiple mechanical properties of Ch-iPSC and chondrocyte constructs shows that using Ch-iPSCs can produce equivalent or better global mechanical properties to chondrocytes. Therefore, iPSC-seeded cartilage constructs could be a promising solution to repair focal cartilage defects. The chondrocyte constructs used in this study have been implanted into humans for clinical trials. Therefore, Ch-iPSC constructs could also be used clinically in place of the current chondrocyte construct.
Collapse
Affiliation(s)
- Jill M Middendorf
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, New York
| | - Nicole Diamantides
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | | | | | | | - Itai Cohen
- Department of Applied Engineering and Physics, Cornell University, Ithaca, New York.,Department of Physics, Cornell University, Ithaca, New York
| | - Lawrence J Bonassar
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, New York.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| |
Collapse
|
29
|
Horne DA, Jones PD, Adams MS, Lotz JC, Diederich CJ. LIPUS far-field exposimetry system for uniform stimulation of tissues in-vitro: development and validation with bovine intervertebral disc cells. Biomed Phys Eng Express 2020; 6:035033. [PMID: 33438678 DOI: 10.1088/2057-1976/ab8b26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Therapeutic Low-intensity Pulsed Ultrasound (LIPUS) has been applied clinically for bone fracture healing and has been shown to stimulate extracellular matrix (ECM) metabolism in numerous soft tissues including intervertebral disc (IVD). In-vitro LIPUS testing systems have been developed and typically include polystyrene cell culture plates (CCP) placed directly on top of the ultrasound transducer in the acoustic near-field (NF). This configuration introduces several undesirable acoustic artifacts, making the establishment of dose-response relationships difficult, and is not relevant for targeting deep tissues such as the IVD, which may require far-field (FF) exposure from low frequency sources. The objective of this study was to design and validate an in-vitro LIPUS system for stimulating ECM synthesis in IVD-cells while mimicking attributes of a deep delivery system by delivering uniform, FF acoustic energy while minimizing reflections and standing waves within target wells, and unwanted temperature elevation within target samples. Acoustic field simulations and hydrophone measurements demonstrated that by directing LIPUS energy at 0.5, 1.0, or 1.5 MHz operating frequency, with an acoustic standoff in the FF (125-350 mm), at 6-well CCP targets including an alginate ring spacer, uniform intensity distributions can be delivered. A custom FF LIPUS system was fabricated and demonstrated reduced acoustic intensity field heterogeneity within CCP-wells by up to 93% compared to common NF configurations. When bovine IVD cells were exposed to LIPUS (1.5 MHz, 200 μs pulse, 1 kHz pulse frequency, and ISPTA = 120 mW cm-2) using the FF system, sample heating was minimal (+0.81 °C) and collagen content was increased by 2.6-fold compared to the control and was equivalent to BMP-7 growth factor treatment. The results of this study demonstrate that FF LIPUS exposure increases collagen content in IVD cells and suggest that LIPUS is a potential noninvasive therapeutic for stimulating repair of tissues deep within the body such as the IVD.
Collapse
Affiliation(s)
- Devante A Horne
- Department of Orthopaedic Surgery, University of California, San Francisco, United States of America. The UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, and University of California, San Francisco, United States of America. Thermal Therapy Research Group, Radiation Oncology Department, University of California, San Francisco, United States of America
| | | | | | | | | |
Collapse
|
30
|
Comparative Study of Electrospun Scaffolds Containing Native GAGs and a GAG Mimetic for Human Mesenchymal Stem Cell Chondrogenesis. Ann Biomed Eng 2020; 48:2040-2052. [DOI: 10.1007/s10439-020-02499-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/24/2020] [Indexed: 12/20/2022]
|
31
|
Middendorf JM, Dugopolski C, Kennedy S, Blahut E, Cohen I, Bonassar LJ. Heterogeneous matrix deposition in human tissue engineered cartilage changes the local shear modulus and resistance to local construct buckling. J Biomech 2020; 105:109760. [PMID: 32276782 DOI: 10.1016/j.jbiomech.2020.109760] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/05/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022]
Abstract
Human tissue engineered cartilage is a promising solution for focal cartilage defects, but these constructs do not have the same local mechanical properties as native tissue. Most clinically relevant engineered cartilage constructs seed human chondrocytes onto a collagen scaffold, which buckles at low loads and strains. This buckling creates local regions of high strain that could cause cell death and damage the engineered tissue. Since human tissue engineered cartilage is commonly grown in-vivo prior to implantation, new matrix deposition could improve the local implant mechanics and prevent local tissue buckling. However, the relationship between local biochemical composition and the local mechanics or local buckling probability has never been quantified. Therefore, this study correlated the local biochemical composition of human tissue engineered cartilage constructs using Fourier transform infrared spectroscopy (FTIR) with the local shear modulus and local buckling probability. The local shear modulus and local buckling probability were obtained using a confocal elastography technique. The local shear modulus increased with increases in local aggrecan content in the interior region (inside the scaffold). A minimum amount of aggrecan was required to prevent local construct buckling at physiologic strains. Since the original scaffold was primarily composed of collagen, increases in collagen content due to new matrix deposition was minimal and had little effect on the mechanical properties. Thus, we concluded that aggrecan deposition inside the scaffold pores is the most effective way to improve the mechanical function and prevent local tissue damage in human tissue engineered cartilage constructs.
Collapse
Affiliation(s)
- Jill M Middendorf
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, NY, USA
| | | | | | | | - Itai Cohen
- Physics, Cornell University, Ithaca, NY, USA
| | - Lawrence J Bonassar
- Sibley School of Mechanical Engineering, Cornell University, Ithaca, NY, USA; Meinig School of Biomedical Engineering Cornell University, Ithaca, NY, USA.
| |
Collapse
|
32
|
Sustained release of TGF-β 3 from polysaccharide nanoparticles induces chondrogenic differentiation of human mesenchymal stromal cells. Colloids Surf B Biointerfaces 2020; 189:110843. [PMID: 32044676 DOI: 10.1016/j.colsurfb.2020.110843] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 11/24/2022]
Abstract
Medical treatment of certain diseases and biomedical implants are tending to use delivery systems on the nanoscale basis for biologically active factors including drugs (e. g. antibiotics) or growth factors. Nanoparticles are a useful tool to deliver bioactive substances of different chemical nature directly to the site where it is required in the patient. Here we developed three innovative delivery systems based on different polysaccharides in order to induce a sustained release of TGF-β3 to mediate chondrogenesis of human mesenchymal stromal cells. We were able to encapsulate the protein into nanoparticles and subsequently release TGF-β3 from these particles. The protein was still active and was able to induce chondrogenic differentiation of human mesenchymal stromal cells.
Collapse
|
33
|
McCorry MC, Kim J, Springer NL, Sandy J, Plaas A, Bonassar LJ. Regulation of proteoglycan production by varying glucose concentrations controls fiber formation in tissue engineered menisci. Acta Biomater 2019; 100:173-183. [PMID: 31546030 DOI: 10.1016/j.actbio.2019.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/30/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022]
Abstract
Fibrillar collagens are highly prevalent in the extracellular matrix of all connective tissues and therefore commonly used as a biomaterial in tissue engineering applications. In the native environment, collagen fibers are arranged in a complex hierarchical structure that is often difficult to recreate in a tissue engineered construct. Small leucine rich proteoglycans as well as hyaluronan binding proteoglycans, aggrecan and versican, have been implicated in regulating fiber formation. In this study, we modified proteoglycan production in vitro by altering culture medium glucose concentrations (4500, 1000, 500, 250, and 125 mg/L), and evaluated its effect on the formation of collagen fibers inside tissue engineered meniscal constructs. Reduction of extracellular glucose resulted in a dose dependent decrease in total sulfated glycosaminoglycan (GAG) production, but minimal decreases of decorin and biglycan. However, fibromodulin doubled in production between 125 and 4500 mg/L glucose concentration. A peak in fiber formation was observed at 500 mg/L glucose concentration and corresponded with reductions in total GAG production. Fiber formation reduction at 125 and 250 mg/L glucose concentrations are likely due to changes in metabolic activity associated with a limited supply of glucose. These results point to proteoglycan production as a means to manipulate fiber architecture in tissue engineered constructs. STATEMENT OF SIGNIFICANCE: Fibrillar collagens are highly prevalent in the extracellular matrix of all connective tissues; however achieving appropriate assembly and organization of collagen fibers in engineered connective tissues is a persistent challenge. Proteoglycans have been implicated in regulating collagen fiber organization both in vivo and in vitro, however little is known about methods to control proteoglycan production and the subsequent fiber organization in tissue engineered menisci. Here, we show that media glucose content can be optimized to control proteoglycan production and collagen fiber assembly, with optimal collagen fiber assembly occurring at sub-physiologic levels of glucose.
Collapse
|
34
|
Neidlin M, Chantzi E, Macheras G, Gustafsson MG, Alexopoulos LG. An ex vivo tissue model of cartilage degradation suggests that cartilage state can be determined from secreted key protein patterns. PLoS One 2019; 14:e0224231. [PMID: 31634377 PMCID: PMC6802827 DOI: 10.1371/journal.pone.0224231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022] Open
Abstract
The pathophysiology of osteoarthritis (OA) involves dysregulation of anabolic and catabolic processes associated with a broad panel of proteins that ultimately lead to cartilage degradation. An increased understanding about these protein interactions with systematic in vitro analyses may give new ideas regarding candidates for treatment of OA related cartilage degradation. Therefore, an ex vivo tissue model of cartilage degradation was established by culturing tissue explants with bacterial collagenase II. Responses of healthy and degrading cartilage were analyzed through protein abundance in tissue supernatant with a 26-multiplex protein profiling assay, after exposing the samples to a panel of 55 protein stimulations present in synovial joints of OA patients. Multivariate data analysis including exhaustive pairwise variable subset selection identified the most outstanding changes in measured protein secretions. MMP9 response to stimulation was outstandingly low in degrading cartilage and there were several protein pairs like IFNG and MMP9 that can be used for successful discrimination between degrading and healthy samples. The discovered changes in protein responses seem promising for accurate detection of degrading cartilage. The ex vivo model seems interesting for drug discovery projects related to cartilage degradation, for example when trying to uncover the unknown interactions between secreted proteins in healthy and degrading tissues.
Collapse
Affiliation(s)
- Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| | - Efthymia Chantzi
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
35
|
Torzilli PA, Azimulla A. Ultraviolet light (365 nm) transmission properties of articular cartilage as a function of depth, extracellular matrix, and swelling. J Biomed Mater Res A 2019; 108:327-339. [PMID: 31622534 DOI: 10.1002/jbm.a.36819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/22/2019] [Accepted: 09/10/2019] [Indexed: 11/06/2022]
Abstract
Current tissue engineering approaches for treatment of injured or diseased articular cartilage use ultraviolet light (UV) for in situ photopolymerization of biomaterials to fill chondral and osteochondral defects as well as resurfacing, stiffening and bonding the extracellular matrix and tissue interfaces. The most commonly used UV light wavelength is UVA 365 nm, the least cytotoxic and deepest penetrating. However, little information is available on the transmission of UVA 365 nm light through the cartilage matrix. In the present study, 365 nm UV light transmission was measured as a function of depth through 100 μm thick slices of healthy articular cartilage removed from mature bovine knees. Transmission properties were measured in normal (Native) cartilage and after swelling equilibration in phosphate-buffered saline (Swollen). Single-factor and multiple linear regression analyses were performed to determine depth-dependencies between the effective attenuation coefficients and proteoglycan, collagen and water contents. For both cartilages, a significant depth-dependency was found for the effective attenuation coefficients, being highest at the articular surface (superficial zone) and decreasing with depth. The effective attenuation coefficients for full-thickness cartilages were approximately a third lower than the total attenuation coefficients calculated from the individual slices. Analysis of absorption and scattering effects due to the ECM and chondrocytes found that UV light scatter coefficients were ∼10 times greater than absorption coefficients. The greater transmittance of UV light through the thicker cartilage was attributed to the collagen within the ECM causing significant backscatter forward reflectance.
Collapse
Affiliation(s)
- Peter A Torzilli
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York.,Physiology, Biophysics and System Biology Program,Weill Medical College of Cornell University, New York, New York.,Department of Biomedical Engineering, City College of New York, New York, New York
| | - Abidally Azimulla
- Department of Biomedical Engineering, City College of New York, New York, New York
| |
Collapse
|
36
|
Jiang EY, Sloan SR, Wipplinger C, Kirnaz S, Härtl R, Bonassar LJ. Proteoglycan removal by chondroitinase ABC improves injectable collagen gel adhesion to annulus fibrosus. Acta Biomater 2019; 97:428-436. [PMID: 31425894 DOI: 10.1016/j.actbio.2019.08.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Intervertebral disc (IVD) herniations are currently treated with interventions that leave the IVD with persistent lesions prone to further herniations. Annulus fibrosus (AF) repair has become of interest as a method to seal defects in the IVD and prevent reherniation, but this requires strong adhesion of the implanted biomaterial to the native AF tissue. Our group has previously developed a high-density collagen (HDC) gel for AF repair and tested its efficacy in vivo, but its adhesion to the AF could be improved. Increased cell adhesion to cartilage has previously been reported through chondroitinase ABC (ChABC) digestion, which removes proteoglycans and increases access to cell binding motifs. Such approaches could also increase biomaterial adhesion to tissue, but the effects of ChABC digestion on AF have yet to be investigated. In this study, ovine AF tissue was digested with either 10 U/mL ChABC or saline for up to 10 min and the effect of this treatment on collagen adhesion between AF tissue samples was investigated by histology and mechanical testing in a lap-shear configuration. ChABC digestion removed proteoglycans within the AF in a time-dependent fashion and enhanced adhesion of the HDC gel to the AF. ChABC digestion increased the elastic toughness and total shear energy of the HDC gel-AF interface by 88% and 46% respectively. ChABC treatment enhanced the adhesion of the HDC gel to the AF without significantly decreasing native AF cell viability. Thus, ChABC digestion is a viable method to improve adhesion of biomaterials for AF repair. STATEMENT OF SIGNIFICANCE: Intervertebral disc herniations are currently treated with interventions that leave persistent lesions in the annulus fibrosus that are prone to further herniations. Annular repair is a promising method to seal lesions and prevent reherniation, but requires strong adhesion of the implanted biomaterial to native annulus fibrosus. Since large proteoglycans like aggrecan occupy regions of the extracellular matrix between collagen fibers in the annulus fibrosus, we hypothesized that removing proteoglycans via chondroitinase digestion would increase the adhesion of annular repair hydrogels. This investigation demonstrated that chondroitinase removed proteoglycans within annulus fibrosus tissue, enhanced the interaction of an injected collagen gel with the native tissue, and mechanically improved adhesion between the collagen gel and annulus fibrosus. This is the first study of its kind to evaluate the biochemical and mechanical effects of short-term chondroitinase digestion on annulus fibrosus tissue.
Collapse
|
37
|
Chastain KS, Stoker AM, Bozynski CC, Leary EV, Cook JL. Metabolic responses of meniscal tissue to focal collagenase degeneration. Connect Tissue Res 2019; 61:349-359. [PMID: 31542969 DOI: 10.1080/03008207.2019.1666112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: The objective of this study was to determine the responses of normal meniscus to collagenase activity. It was hypothesized that meniscal explants exposed to collagenase would significantly increase release of pro-inflammatory cytokines and degradative enzymes, in a dose-dependent manner, compared to control.Methods: Menisci were harvested from adult dogs (n = 6) euthanized for reasons unrelated to this study. Meniscal explants were created from the central portion of lateral and medial meniscus. Explants were injected with 100 µl collagenase at a concentration of 50 µg/ml, 5 µg/ml, or 0 µg/ml of collagenase. Explants were cultured for 12 days, and media were changed and collected every 3 days for biomarker analyses. Differences among collagenase concentrations were determined by a three factor ANOVA with adjustment for multiple comparisons, with pre-adjustment statistical significance set at p < 0.05.Results: When data from all explants were compared, the 50 µg group released significantly higher IL-6 and PGE2, and the 5 µg group released significantly higher levels of MMP-3 and CTX-II compared to the 0 µg group. Explants from the medial meniscus released significantly more MMP-1, MMP-2, MMP-3, and MMP-13 in response to stimulation with 5 µg/ml of collagenase compared to explants from the lateral meniscus.Discussion: The data from this study indicate that in response to localized degradative enzyme activity, the meniscus increases the release of pro-inflammatory and degradative biomarkers in a dose-dependent manner. Further, these data indicate potential differences in metabolic responses of lateral versus medial menisci to collagenase insult.
Collapse
Affiliation(s)
- Kamryn S Chastain
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA.,Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| | - Aaron M Stoker
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA.,Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| | - Chantelle C Bozynski
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA.,Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| | - Emily V Leary
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA.,Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| | - James L Cook
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA.,Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| |
Collapse
|
38
|
Abstract
PURPOSE The significant shortcomings associated with current autologous reconstructive options for auricular deformities have inspired great interest in a tissue engineering solution. A major obstacle in the engineering of human auricular cartilage is the availability of sufficient autologous human chondrocytes. A clinically obtainable amount of auricular cartilage tissue (ie, 1 g) only yields approximately 10 million cells, where 25 times this amount is needed for the fabrication of a full-scale pediatric ear. It is thought that repeated passaging of chondrocytes leads to dedifferentiation and loss of the chondrogenic potential. However, little to no data exist regarding the ideal number of times that human auricular chondrocytes (HAuCs) can be passaged in a manner that maximizes the cellular expansion while minimizing dedifferentiation. METHODS Human auricular chondrocytes were isolated from discarded otoplasty specimens. The HAuCs were then expanded, and cells from passages 3, 4, and 5 were encapsulated into discs 8 mm in diameter made from type I collagen hydrogels with a cell density of 25 million cells/mL. The constructs were implanted subcutaneously in the dorsa of nude mice and harvested after 1 and 3 months for analysis. RESULTS Constructs containing passages 3, 4, and 5 chondrocytes all maintained their original cylindrical geometry. After 3 months in vivo, the diameters of the P3, P4, and P5 discs were 69 ± 9%, 67 ± 10%, and 73 ± 15% of their initial diameter, respectively. Regardless of the passage number, all constructs developed a glossy white cartilaginous appearance, similar to native auricular cartilage. Histologic analysis demonstrated development of an organized perichondrium composed of collagen, a rich proteoglycan matrix, cellular lacunae, and a dense elastin fibrin network by Safranin-O and Verhoeff stain. Biochemical analysis confirmed similar amounts of proteoglycan and hydroxyproline content in late passage constructs when compared with native auricular cartilage. CONCLUSIONS These data indicate that late passage HAuCs (up to passage 5) form elastic cartilage that is histologically, biochemically, and biomechanically similar to native human elastic cartilage and have the potential to be used for auricular cartilage engineering.
Collapse
|
39
|
Corrêa-Ferreira ML, Viudes EB, de Magalhães PM, Paixão de Santana Filho A, Sassaki GL, Pacheco AC, de Oliveira Petkowicz CL. Changes in the composition and structure of cell wall polysaccharides from Artemisia annua in response to salt stress. Carbohydr Res 2019; 483:107753. [DOI: 10.1016/j.carres.2019.107753] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
|
40
|
Diamantides N, Dugopolski C, Blahut E, Kennedy S, Bonassar LJ. High density cell seeding affects the rheology and printability of collagen bioinks. Biofabrication 2019; 11:045016. [PMID: 31342915 DOI: 10.1088/1758-5090/ab3524] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
An advantage of bioprinting is the ability to incorporate cells into the hydrogel bioink allowing for precise control over cell placement within a construct. Previous work found that the printability of collagen bioinks is highly dependent on their rheological properties. The effect of cell density on collagen rheological properties and, therefore, printability has not been assessed. Therefore, the objective of this study was to determine the effects of incorporating cells on the rheology and printability of collagen bioinks. Primary chondrocytes, at densities relevant to cartilage tissue engineering (up to 100 × 106 cells ml-1), were incorporated into 8 mg ml-1 collagen bioinks. Bioink rheological properties before, during, and after gelation as well as printability were assessed. Cell-laden printed constructs were also cultured for up to 14 d to assess longer-term cell behavior. The addition of cells resulted in an increase in the storage modulus and viscosity of the collagen before gelation. However, the storage modulus after gelation and the rate of gelation decreased with increasing cell density. Theoretical models were compared to the rheological data to suggest frameworks that could be used to predict the rheological properties of cell-laden bioinks. Printability testing showed that improved printability could be achieved with higher cell densities. Fourteen-day culture studies showed that the printing process had no adverse effects on the viability or function of printed cells. Overall, this study shows that collagen bioinks are conducive to bioprinting with a wide range of cell densities while maintaining high printability and chondrocyte viability and function.
Collapse
Affiliation(s)
- Nicole Diamantides
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | | | | | | | | |
Collapse
|
41
|
Qu D, Zhu JP, Childs HR, Lu HH. Nanofiber-based transforming growth factor-β3 release induces fibrochondrogenic differentiation of stem cells. Acta Biomater 2019; 93:111-122. [PMID: 30862549 DOI: 10.1016/j.actbio.2019.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
Fibrocartilage is typically found in regions subject to complex, multi-axial loads and plays a critical role in musculoskeletal function. Mesenchymal stem cell (MSC)-mediated fibrocartilage regeneration may be guided by administration of appropriate chemical and/or physical cues, such as by culturing cells on polymer nanofibers in the presence of the chondrogenic growth factor TGF-β3. However, targeted delivery and maintenance of effective local factor concentrations remain challenges for implementation of growth factor-based regeneration strategies in clinical settings. Thus, the objective of this study was to develop and optimize the bioactivity of a biomimetic nanofiber scaffold system that enables localized delivery of TGF-β3. To this end, we fabricated TGF-β3-releasing nanofiber meshes that provide sustained growth factor delivery and demonstrated their potential for guiding synovium-derived stem cell (SDSC)-mediated fibrocartilage regeneration. TGF-β3 delivery enhanced cell proliferation and synthesis of relevant fibrocartilaginous matrix in a dose-dependent manner. By designing a scaffold that eliminates the need for exogenous or systemic growth factor administration and demonstrating that fibrochondrogenesis requires a lower growth factor dose compared to previously reported, this study represents a critical step towards developing a clinical solution for regeneration of fibrocartilaginous tissues. STATEMENT OF SIGNIFICANCE: Fibrocartilage is a tissue that plays a critical role throughout the musculoskeletal system. However, due to its limited self-healing capacity, there is a significant unmet clinical need for more effective approaches for fibrocartilage regeneration. We have developed a nanofiber-based scaffold that provides both the biomimetic physical cues, as well as localized delivery of the chemical factors needed to guide stem cell-mediated fibrocartilage formation. Specifically, methods for fabricating TGF-β3-releasing nanofibers were optimized, and scaffold-mediated TGF-β3 delivery enhanced cell proliferation and synthesis of fibrocartilaginous matrix, demonstrating for the first time, the potential for nanofiber-based TGF-β3 delivery to guide stem cell-mediated fibrocartilage regeneration. This nanoscale delivery platform represents an exciting new strategy for fibrocartilage regeneration.
Collapse
Affiliation(s)
- Dovina Qu
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace Building, MC 8904, 1210 Amsterdam Avenue, New York, NY 10027, United States
| | - Jennifer P Zhu
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace Building, MC 8904, 1210 Amsterdam Avenue, New York, NY 10027, United States
| | - Hannah R Childs
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace Building, MC 8904, 1210 Amsterdam Avenue, New York, NY 10027, United States
| | - Helen H Lu
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace Building, MC 8904, 1210 Amsterdam Avenue, New York, NY 10027, United States.
| |
Collapse
|
42
|
Zhou X, Tang X, Long R, Wang S, Wang P, Cai D, Liu Y. The Influence of bFGF on the Fabrication of Microencapsulated Cartilage Cells under Different Shaking Modes. Polymers (Basel) 2019; 11:polym11030471. [PMID: 30960455 PMCID: PMC6473345 DOI: 10.3390/polym11030471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/28/2019] [Accepted: 03/06/2019] [Indexed: 12/02/2022] Open
Abstract
Cell encapsulation in hydrogels has been extensively used in cytotherapy, regenerative medicine, 3D cell culture, and tissue engineering. Herein, we fabricated microencapsulated cells through microcapsules loaded with C5.18 chondrocytes alginate/chitosan prepared by a high-voltage electrostatic method. Under optimized conditions, microencapsulated cells presented uniform size distribution, good sphericity, and a smooth surface with different cell densities. The particle size distribution was determined at 150–280 μm, with an average particle diameter of 220 μm. The microencapsulated cells were cultured under static, shaking, and 3D micro-gravity conditions with or without bFGF (basic fibroblast growth factor) treatment. The quantified detection (cell proliferation detection and glycosaminoglycan (GAG)/type II collagen (Col-II)) content was respectively determined by cell counting kit-8 assay (CCK-8) and dimethylmethylene blue (DMB)/Col-II secretion determination) and qualitative detection (acridine orange/ethidium bromide, hematoxylin-eosin, alcian blue, safranin-O, and immunohistochemistry staining) of these microencapsulated cells were evaluated. Results showed that microencapsulated C5.18 cells under three-dimensional microgravity conditions promoted cells to form large cell aggregates within 20 days by using bFGF, which provided the possibility for cartilage tissue constructs in vitro. It could be found from the cell viability (cell proliferation) and synthesis (content of GAG and Col-II) results that microencapsulated cells had a better cell proliferation under 3D micro-gravity conditions using bFGF than under 2D conditions (including static and shaking conditions). We anticipate that these results will be a benefit for the design and construction of cartilage regeneration in future tissue engineering applications.
Collapse
Affiliation(s)
- Xia Zhou
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Xiaolin Tang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ruimin Long
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China.
| | - Shibin Wang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China.
- Institutes of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Pei Wang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Duanhua Cai
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Yuangang Liu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China.
- Institutes of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
43
|
Hung BP, Harvestine JN, Saiz AM, Gonzalez-Fernandez T, Sahar DE, Weiss ML, Leach JK. Defining hydrogel properties to instruct lineage- and cell-specific mesenchymal differentiation. Biomaterials 2019; 189:1-10. [PMID: 30384124 PMCID: PMC6237198 DOI: 10.1016/j.biomaterials.2018.10.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 02/06/2023]
Abstract
The maintenance and direction of stem cell lineage after implantation remains challenging for clinical translation. Aggregation and encapsulation into instructive biomaterials after preconditioning can bolster retention of differentiated phenotypes. Since these procedures do not depend on cell type or lineage, we hypothesized we could use a common, tunable platform to engineer formulations that retain and enhance multiple lineages from different cell populations. To test this, we varied alginate stiffness and adhesive ligand content, then encapsulated spheroids of varying cellularity. We used Design-of-Experiments to determine the effect of these parameters and their interactions on phenotype retention. The combination of parameters leading to maximal differentiation varied with lineage and cell type, inducing a 2-4-fold increase over non-optimized levels. Phenotype was also retained for 4 weeks in a murine subcutaneous model. This widely applicable approach can facilitate translation of cell-based therapies by instructing phenotype in situ without prolonged induction or costly growth factors.
Collapse
Affiliation(s)
- Ben P Hung
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Jenna N Harvestine
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Augustine M Saiz
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, USA
| | | | - David E Sahar
- Department of Surgery, Division of Plastic Surgery, UC Davis Health, Sacramento, CA 95817, USA
| | - Mark L Weiss
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, USA.
| |
Collapse
|
44
|
DiDomenico CD, Kaghazchi A, Bonassar LJ. Measurement of local diffusion and composition in degraded articular cartilage reveals the unique role of surface structure in controlling macromolecular transport. J Biomech 2018; 82:38-45. [PMID: 30385000 DOI: 10.1016/j.jbiomech.2018.10.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/05/2018] [Accepted: 10/17/2018] [Indexed: 12/23/2022]
Abstract
Developing effective therapeutics for osteoarthritis (OA) necessitates that such molecules can reach and target chondrocytes within articular cartilage. However, predicting how well very large therapeutic molecules diffuse through cartilage is often difficult, and the relationship between local transport mechanics for these molecules and tissue heterogeneities in the tissue is still unclear. In this study, a 150 kDa antibody diffused through the articular surface of healthy and enzymatically degraded cartilage, which enabled the calculation of local diffusion mechanics in tissue with large compositional variations. Local cartilage composition and structure was quantified with Fourier transform infrared (FTIR) spectroscopy and second harmonic generation (SHG) imaging techniques. Overall, both local concentrations of aggrecan and collagen were correlated to local diffusivities for both healthy and surface-degraded samples (0.3 > R2 < 0.9). However, samples that underwent surface degradation by collagenase exhibited stronger correlations (R2 > 0.75) compared to healthy samples (R2 < 0.46), suggesting that the highly aligned collagen at the surface of cartilage acts as a barrier to macromolecular transport.
Collapse
Affiliation(s)
- Chris D DiDomenico
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Aydin Kaghazchi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States; Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
45
|
Cohen BP, Bernstein JL, Morrison KA, Spector JA, Bonassar LJ. Tissue engineering the human auricle by auricular chondrocyte-mesenchymal stem cell co-implantation. PLoS One 2018; 13:e0202356. [PMID: 30356228 PMCID: PMC6200177 DOI: 10.1371/journal.pone.0202356] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/01/2018] [Indexed: 01/21/2023] Open
Abstract
Children suffering from microtia have few options for auricular reconstruction. Tissue engineering approaches attempt to replicate the complex anatomy and structure of the ear with autologous cartilage but have been limited by access to clinically accessible cell sources. Here we present a full-scale, patient-based human ear generated by implantation of human auricular chondrocytes and human mesenchymal stem cells in a 1:1 ratio. Additional disc construct surrogates were generated with 1:0, 1:1, and 0:1 combinations of auricular chondrocytes and mesenchymal stem cells. After 3 months in vivo, monocellular auricular chondrocyte discs and 1:1 disc and ear constructs displayed bundled collagen fibers in a perichondrial layer, rich proteoglycan deposition, and elastin fiber network formation similar to native human auricular cartilage, with the protein composition and mechanical stiffness of native tissue. Full ear constructs with a 1:1 cell combination maintained gross ear structure and developed a cartilaginous appearance following implantation. These studies demonstrate the successful engineering of a patient-specific human auricle using exclusively human cell sources without extensive in vitro tissue culture prior to implantation, a critical step towards the clinical application of tissue engineering for auricular reconstruction.
Collapse
Affiliation(s)
- Benjamin P Cohen
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Jaime L Bernstein
- Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, United States of America
| | - Kerry A Morrison
- Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, United States of America
| | - Jason A Spector
- Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, United States of America
| | - Lawrence J Bonassar
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
46
|
Jenkins TL, Meehan S, Pourdeyhimi B, Little D. * Meltblown Polymer Fabrics as Candidate Scaffolds for Rotator Cuff Tendon Tissue Engineering. Tissue Eng Part A 2018; 23:958-967. [PMID: 28816097 DOI: 10.1089/ten.tea.2016.0470] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Various biomaterial technologies are promising for tissue engineering, including electrospinning, but commercial scale-up of throughput is difficult. The goal of the study was to evaluate meltblown fabrics as candidate scaffolds for rotator cuff tendon tissue engineering. Meltblown poly(lactic acid) fabrics were produced with several polymer crystallinities and airflow velocities [500(low), 900(medium) or 1400(high) m3air/h/m fabric]. Fiber diameter, alignment, and baseline bidirectional tensile mechanical properties were evaluated. Attachment and spreading of human adipose-derived stem cells (hASCs) were evaluated over 3 days immediately following seeding. After initial screening, the fabric with the greatest Young's modulus and yield stress was selected for 28-day in vitro culture and for evaluation of tendon-like extracellular matrix production and development of mechanical properties. As expected, airflow velocity of the polymer during meltblowing demonstrated an inverse relationship with fiber diameter. All fabrics exhibited fiber alignment parallel to the direction of collector rotation. All fabrics demonstrated mechanical anisotropy at baseline. Cells attached, proliferated, and spread on all fabrics over the initial three-day culture period. Consistent with the observed loss of integrity of the unseeded biomaterial, hASC-seeded scaffolds demonstrated a significant decrease in Young's modulus over 28 days of culture. However, dsDNA, sulfated glycosaminoglycan, and collagen content increased significantly over 28 days. Histology and polarized light microscopy demonstrated collagen deposition and alignment throughout the thickness of the scaffolds. While fiber diameters approximated an order of magnitude greater than those previously reported for electrospun scaffolds intended for tendon tissue engineering, they were still within the range of collagen fiber diameters found in healthy tendon. The extent of matrix production and alignment was similar to that previously observed for multilayered electrospun scaffolds. While the Young's modulus of scaffolds after 28 days of culture was lower than native rotator cuff tendon, it approximated that reported previously following culture of electrospun scaffolds and was on the same order of magnitude as of current Food and Drug Administration-approved patches for rotator cuff augmentation. Together, these data suggest that with minor polymer and parameter modifications, meltblown scaffolds could provide an economical, high-throughput production alternative method to electrospinning for use in rotator cuff tendon tissue engineering.
Collapse
Affiliation(s)
- Thomas L Jenkins
- 1 Department of Basic Medical Science, Purdue University College of Veterinary Medicine and Department of Biomedical Engineering, Weldon School of Engineering, Purdue University , West Lafayette, Indiana.,2 Department of Orthopaedic Surgery, Duke University , Durham, North Carolina
| | - Sean Meehan
- 2 Department of Orthopaedic Surgery, Duke University , Durham, North Carolina
| | - Behnam Pourdeyhimi
- 3 The Nonwovens Institute, North Carolina State University , Raleigh, North Carolina
| | - Dianne Little
- 1 Department of Basic Medical Science, Purdue University College of Veterinary Medicine and Department of Biomedical Engineering, Weldon School of Engineering, Purdue University , West Lafayette, Indiana.,2 Department of Orthopaedic Surgery, Duke University , Durham, North Carolina
| |
Collapse
|
47
|
Duan W, Chen C, Haque M, Hayes D, Lopez MJ. Polymer-mineral scaffold augments in vivo equine multipotent stromal cell osteogenesis. Stem Cell Res Ther 2018. [PMID: 29523214 PMCID: PMC5845133 DOI: 10.1186/s13287-018-0790-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Use of bioscaffolds to direct osteogenic differentiation of adult multipotent stromal cells (MSCs) without exogenous proteins is a contemporary approach to bone regeneration. Identification of in vivo osteogenic contributions of exogenous MSCs on bioscaffolds after long-term implantation is vital to understanding cell persistence and effect duration. Methods This study was designed to quantify in vivo equine MSC osteogenesis on synthetic polymer scaffolds with distinct mineral combinations 9 weeks after implantation in a murine model. Cryopreserved, passage (P)1, equine bone marrow-derived MSCs (BMSC) and adipose tissue-derived MSCs (ASC) were culture expanded to P3 and immunophenotyped with flow cytometry. They were then loaded by spinner flask on to scaffolds composed of tricalcium phosphate (TCP)/hydroxyapatite (HA) (40:60; HT), polyethylene glycol (PEG)/poly-l-lactic acid (PLLA) (60:40; GA), or PEG/PLLA/TCP/HA (36:24:24:16; GT). Scaffolds with and without cells were maintained in static culture for up to 21 days or implanted subcutaneously in athymic mice that were radiographed every 3 weeks up to 9 weeks. In vitro cell viability and proliferation were determined. Explant composition (double-stranded (ds)DNA, collagen, sulfated glycosaminoglycan (sGAG), protein), equine and murine osteogenic target gene expression, microcomputed tomography (μCT) mineralization, and light microscopic structure were assessed. Results The ASC and BMSC number increased significantly in HT constructs between 7 and 21 days of culture, and BMSCs increased similarly in GT constructs. Radiographic opacity increased with time in GT-BMSC constructs. Extracellular matrix (ECM) components and dsDNA increased significantly in GT compared to HT constructs. Equine and murine osteogenic gene expression was highest in BMSC constructs with mineral-containing scaffolds. The HT constructs with either cell type had the highest mineral deposition based on μCT. Regardless of composition, scaffolds with cells had more ECM than those without, and osteoid was apparent in all BMSC constructs. Conclusions In this study, both exogenous and host MSCs appear to contribute to in vivo osteogenesis. Addition of mineral to polymer scaffolds enhances equine MSC osteogenesis over polymer alone, but pure mineral scaffold provides superior osteogenic support. These results emphasize the need for bioscaffolds that provide customized osteogenic direction of both exo- and endogenous MSCs for the best regenerative potential.
Collapse
Affiliation(s)
- Wei Duan
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Cong Chen
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Masudul Haque
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Daniel Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
48
|
Pleumeekers MM, Nimeskern L, Koevoet JLM, Karperien M, Stok KS, van Osch GJVM. Trophic effects of adipose-tissue-derived and bone-marrow-derived mesenchymal stem cells enhance cartilage generation by chondrocytes in co-culture. PLoS One 2018; 13:e0190744. [PMID: 29489829 PMCID: PMC5830031 DOI: 10.1371/journal.pone.0190744] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 12/10/2017] [Indexed: 01/22/2023] Open
Abstract
AIMS Combining mesenchymal stem cells (MSCs) and chondrocytes has great potential for cell-based cartilage repair. However, there is much debate regarding the mechanisms behind this concept. We aimed to clarify the mechanisms that lead to chondrogenesis (chondrocyte driven MSC-differentiation versus MSC driven chondroinduction) and whether their effect was dependent on MSC-origin. Therefore, chondrogenesis of human adipose-tissue-derived MSCs (hAMSCs) and bone-marrow-derived MSCs (hBMSCs) combined with bovine articular chondrocytes (bACs) was compared. METHODS hAMSCs or hBMSCs were combined with bACs in alginate and cultured in vitro or implanted subcutaneously in mice. Cartilage formation was evaluated with biochemical, histological and biomechanical analyses. To further investigate the interactions between bACs and hMSCs, (1) co-culture, (2) pellet, (3) Transwell® and (4) conditioned media studies were conducted. RESULTS The presence of hMSCs-either hAMSCs or hBMSCs-increased chondrogenesis in culture; deposition of GAG was most evidently enhanced in hBMSC/bACs. This effect was similar when hMSCs and bAC were combined in pellet culture, in alginate culture or when conditioned media of hMSCs were used on bAC. Species-specific gene-expression analyses demonstrated that aggrecan was expressed by bACs only, indicating a predominantly trophic role for hMSCs. Collagen-10-gene expression of bACs was not affected by hBMSCs, but slightly enhanced by hAMSCs. After in-vivo implantation, hAMSC/bACs and hBMSC/bACs had similar cartilage matrix production, both appeared stable and did not calcify. CONCLUSIONS This study demonstrates that replacing 80% of bACs by either hAMSCs or hBMSCs does not influence cartilage matrix production or stability. The remaining chondrocytes produce more matrix due to trophic factors produced by hMSCs.
Collapse
Affiliation(s)
- M. M. Pleumeekers
- Department of Otorhinolaryngology, Head and Neck surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - L. Nimeskern
- Institute for Biomechanics, ETH, Zürich, Switzerland
| | - J. L. M. Koevoet
- Department of Otorhinolaryngology, Head and Neck surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - M. Karperien
- Department of Tissue Regeneration, MIRA-institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - K. S. Stok
- Institute for Biomechanics, ETH, Zürich, Switzerland
| | - G. J. V. M. van Osch
- Department of Otorhinolaryngology, Head and Neck surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
49
|
Mallick SP, Singh BN, Rastogi A, Srivastava P. Design and evaluation of chitosan/poly(l-lactide)/pectin based composite scaffolds for cartilage tissue regeneration. Int J Biol Macromol 2018; 112:909-920. [PMID: 29438752 DOI: 10.1016/j.ijbiomac.2018.02.049] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/14/2017] [Accepted: 02/08/2018] [Indexed: 01/01/2023]
Abstract
Poor regenerative potential of cartilage tissue due to the avascular nature and lack of supplementation of reparative cells impose an important challenge in recent medical practice towards development of artificial extracellular matrix with enhanced neo-cartilage tissue regeneration potential. Chitosan (CH), poly (l-lactide) (PLLA), and pectin (PC) compositions were tailored to generate polyelectrolyte complex based porous scaffolds using freeze drying method and crosslinked by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC), N-hydroxysuccinimide (NHS) solution containing chondroitin sulfate (CS) to mimic the composition as well as architecture of the cartilage extracellular matrix (ECM). The physical, chemical, thermal, and mechanical behaviors of developed scaffolds were done. The scaffolds were porous with homogeneous pore structure with pore size 49-170μm and porosities in the range of 79 to 84%. Fourier transform infrared study confirmed the presence of polymers (CH, PLLA and PC) within the scaffolds. The crystallinity of the scaffold was examined by the X-ray diffraction studies. Furthermore, scaffold shows suitable swelling property, moderate biodegradation and hemocompatibility in nature and possess suitable mechanical strength for cartilage tissue regeneration. MTT assay, GAG content, and attachment of chondrocyte confirmed the regenerative potential of the cell seeded scaffold. The histopathological analysis defines the suitability of scaffold for cartilage tissue regeneration.
Collapse
Affiliation(s)
- Sarada Prasanna Mallick
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Bhisham Narayan Singh
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Amit Rastogi
- Department of Orthopedics, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Pradeep Srivastava
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India.
| |
Collapse
|
50
|
Effect of Cyclic Dynamic Compressive Loading on Chondrocytes and Adipose-Derived Stem Cells Co-Cultured in Highly Elastic Cryogel Scaffolds. Int J Mol Sci 2018; 19:ijms19020370. [PMID: 29373507 PMCID: PMC5855592 DOI: 10.3390/ijms19020370] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/23/2022] Open
Abstract
In this study, we first used gelatin/chondroitin-6-sulfate/hyaluronan/chitosan highly elastic cryogels, which showed total recovery from large strains during repeated compression cycles, as 3D scaffolds to study the effects of cyclic dynamic compressive loading on chondrocyte gene expression and extracellular matrix (ECM) production. Dynamic culture of porcine chondrocytes was studied at 1 Hz, 10% to 40% strain and 1 to 9 h/day stimulation duration, in a mechanical-driven multi-chamber bioreactor for 14 days. From the experimental results, we could identify the optimum dynamic culture condition (20% and 3 h/day) to enhance the chondrocytic phenotype of chondrocytes from the expression of marker (Col I, Col II, Col X, TNF-α, TGF-β1 and IGF-1) genes by quantitative real-time polymerase chain reactions (qRT-PCR) and production of ECM (GAGs and Col II) by biochemical analysis and immunofluorescence staining. With up-regulated growth factor (TGF-β1 and IGF-1) genes, co-culture of chondrocytes with porcine adipose-derived stem cells (ASCs) was employed to facilitate chondrogenic differentiation of ASCs during dynamic culture in cryogel scaffolds. By replacing half of the chondrocytes with ASCs during co-culture, we could obtain similar production of ECM (GAGs and Col II) and expression of Col II, but reduced expression of Col I, Col X and TNF-α. Subcutaneous implantation of cells/scaffold constructs in nude mice after mono-culture (chondrocytes or ASCs) or co-culture (chondrocytes + ASCs) and subject to static or dynamic culture condition in vitro for 14 days was tested for tissue-engineering applications. The constructs were retrieved 8 weeks post-implantation for histological analysis by Alcian blue, Safranin O and Col II immunohistochemical staining. The most abundant ectopic cartilage tissue was found for the chondrocytes and chondrocytes + ASCs groups using dynamic culture, which showed similar neo-cartilage formation capability with half of the chondrocytes replaced by ASCs for co-culture. This combined co-culture/dynamic culture strategy is expected to cut down the amount of donor chondrocytes needed for cartilage-tissue engineering.
Collapse
|